ArticlePDF AvailableLiterature Review

Abstract

The process of creatine synthesis occurs in two steps, catalyzed by L-arginine:glycine amidinotransferase (AGAT) and guanidinoacetate N-methyltransferase (GAMT), which takes place mainly in kidney and liver, respectively. This molecule plays an important energy/pH buffer function in tissues, and to guarantee the maintenance of its total body pool, the lost creatine must be replaced from diet or de novo synthesis. Creatine administration is known to decrease the consumption of S-adenosyl methionine and also reduce the homocysteine production in liver, diminishing fat accumulation and resulting in beneficial effects in fatty liver and non-alcoholic liver disease. Different studies have shown that creatine supplementation could supply brain energy, presenting neuroprotective effects against the encephalopathy induced by hyperammonemia in acute liver failure. Creatine is also taken by many athletes for its ergogenic properties. However, little is known about the adverse effects of creatine supplementation, which are barely described in the literature, with reports of mainly hypothetical effects arising from a small number of scientific publications. Antioxidant effects have been found in several studies, although one of the theories regarding the potential for toxicity from creatine supplementation is that it can increase oxidative stress and potentially form carcinogenic compounds.
Send Ord ers for Reprints to reprints@benthamscience.ae
12 Mini-Reviews in Medicinal Chemistry, 2016, 16, 12-18
Creatine and the Liver: Metabolism and Possible Interactions
R.P. Barcelos1,2, S.T. Stefanello1, J.L. Mauriz2, J. González-Gallego2 and F.A.A. Soares1*
1Departamento de Bioquímica e Biologia Molecular, Centro de Ciências Naturais e Exatas (CCNE),
Universidade Federal de Santa Maria, Santa Maria (UFSM), Rio Grande do Sul, Brasil; 2Institute of
Biomedicine (IBIOMED) and CIBERehd, University of León, León, Spain
Abstract: The process of creatine synthesis occurs in two steps, catalyzed by L-arginine:glycine
amidinotransferase (AGAT) and guanidinoacetate N-methyltransferase (GAMT), which take place
mainly in kidney and liver, respectively. This molecule plays an important energy/pH buffer function
in tissues, and to guarantee the maintenance of its total body pool, the lost creatine must be replaced
from diet or de novo synthesis. Creatine administration is known to decrease the consumption of S-
adenosyl methionine and also reduce the homocysteine production in liver, diminishing fat
accumulation and resulting in beneficial effects in fatty liver and non-alcoholic liver disease. Different studies have shown
that creatine supplementation could supply brain energy, presenting neuroprotective effects against the encephalopathy
induced by hyperammonemia in acute liver failure. Creatine is also taken by many athletes for its ergogenic properties.
However, little is known about the adverse effects of creatine supplementation, which are barely described in the
literature, with reports of mainly hypothetical effects arising from a small number of scientific publications. Antioxidant
effects have been found in several studies, although one of the theories regarding the potential for toxicity from creatine
supplementation is that it can increase oxidative stress and potentially form carcinogenic compounds.
Keywords: Creatine, damage, exercise, kidney, liver.
1. THE LIVER ROLE IN THE SYNTHESIS OF
CREATINE
Creatine occurs naturally in food, especially in meat and
fish [1], although humans on a typical Western diet obtain
about one-half of their creatine by synthesis and one-half
from the diet [2, 3]. However, vegetarians obtain very little
dietary creatine, and the endogenous synthesis is their major
source [4]. Creatine plays an important energy/pH buffer
function in tissues, which continuously require a replacement
of creatine stores through the dietary or body synthesis [5].
The need to synthesize creatine arises from the fact that
in a young 70-kg male, under physiological conditions, there
is a loss of 1-2 g of creatine per day by the spontaneously
and irreversible conversion to creatinine [1, 6], which is
subsequently lost to the urine. Therefore, to guarantee the
maintenance of the total body pool of creatine, the lost
creatine must be replaced from diet or de novo synthesis [2].
The process of creatine biosynthesis occur in two steps
(Fig. 1) that require three amino acids, arginine, glycine
and methionine, and the enzymes L-arginine:glycine
amidinotransferase (AGAT, EC 2.1.4.1) and guanidinoacetate
N-methyltransferase (GAMT, EC 2.1.1.2) [7]. During the
first step, the amidino group from arginine is transferred to
the amino group of glycine, yielding ornithine and
*Address correspondence to this author at the Departamento de Bioquímica
e Biologia Molecular, Centro de Ciências Naturais e Exatas, Universidade
Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil; Tel: +55
55 32209522; Fax: +55 55 3220 8978; E-mail: felix@ufsm.br
guanidinoacetic acid (GAA), reaction catalyzed by the
enzyme AGAT [2, 5, 8]. In the second step, GAMT induces
the GAA methylation, on the original glycine nitrogen, using
S-adenosylmethionine (SAM) as the methyl donor to form S-
adenosylhomocysteine (SAH) and creatine [2, 5, 8]. It has
been suggested that AGAT is a critical control step in the
creatine biosynthesis, and creatine supplementation could
downregulate its expression [2, 9].
There are high activities of AGAT in the kidneys and of
GAMT in the livers of various species [10]. Such tissue
enrichment has suggested that creatine synthesis is an
interorgan process [11]. On the basis mostly of these latter
findings and of the fact that the rate of creatine biosynthesis
is considerably reduced in nephrectomized animals [12-14],
it was postulated, and is still largely accep ted, that the main
route of creatine biosynthesis in mammals involves
formation of GAA, produced by the kidney, released into the
circulation for transportation through the blood and then
methylated to creatine in the liver [11] (Fig. 2).
However, both AGAT and GAMT occur in tissues other
than kidney and liver. In particular, it is known that brain can
synthesize creatine [8]. Since it is known that the blood-brain
barrier is poorly permeable to creatine, the central nervous
system can be considered an autonomous tissue in creatine
synthesis [15]. In mammals, pancreas contains high levels of
both enzymes, whereas livers contain high amounts of
GAMT. On the other way, kidneys of all the species tested
so far express high amounts of AGAT but relatively lower
levels of GAMT. Although livers of cow, pig, monkey, and
F.A.A. Soares
1875-5607/16 $58.00+.00 © 2016 Bentham Science Publishers
Creatine and Liver Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 13
Fig. (1). Creatine biosynthesis pathway. In the first step, L-arginine:glycine amidinotransferase (AGAT) transferes an amidino group from
arginine to the amino group of glycine, yielding ornithine and guanidinoacetic acid (GAA). In the second step, GAA is methylated by
the enzyme guanidinoacetate methyltransferase (GAMT), using S-adenosylmethionine (SAM) as the methyl donor, to form
S-adenosylhomocyestine (SAH) and creatine.
Fig. (2). Creatine biosynthesis as an interorgan process. Guanidinoacetic acid (GAA) synthetized in kidney is released into circulation and
transported to the liver where it is methylated to produce creatine. Synthetized creatine is distributed via bloodstream to the organs of usage
(mainly muscle and brain).
Arginine + Glycine
GAA
Ornithine
LIVER
KIDNEY BLOOD
AGAT
GAA GAA
SAM
SAH
Creatine Creatine
(to other
organs and
tissues)
14 Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 Barcelos et al.
human also have high amounts of AGAT, livers of common
laboratory mammals such as the rat, mouse, dog, cat, and
rabbit are reported to lack AGAT activity [16]. However, the
renal production of GAA in humans appears to represent
only 20% of the daily loss of creatinine, suggesting that
GAA must be synthesized in other tissues [2].
It has been suggested that the entire creatine synthetic
pathway can be formed in the liver, in a situation where
arginine could be acted upon by either arginase to form urea
or by AGAT to form GAA. Ornithine would be formed and
metabolized via the urea cycle enzyme ornithine
transcarbamoylase [17]. Some studies reported an AGAT
mRNA expression in human liver [18]. In support of this
hypothesis, immunohistochemical evidence for the AGAT
protein in the cytosol of rat hepatocytes was found [19].
However, some past failures to measure AGAT activity
under standard assay conditions in rat liver are also reported
in literature [2].
Finally, the creatine-requiring tissues, via a specific
transporter, can take up creatine released from the liver into
circulation [11]. Creatine transporters have been described in
muscle, brain, kidney, and intestine [20, 21]. These are Na+-
and Cl--dependent active transporters, which concentrate
creatine intracellularly [22]. However, such transporters
seem inappropriate for the liver.
AGAT is the critical control step in creatine synthesis.
Literature strongly suggests that the regulation of creatine
biosynthesis is usually through this enzyme [5]. While the
growth hormone upregulates AGAT expression [23, 24], an
increase in serum levels of creatine results in a decrease in
AGAT enzyme activity, enzyme level, and mRNA
expression in rat kidney [25], whereas creatine ingestion by
humans lowers plasma GAA levels, which is consistent with
the downregulation of AGAT activity [26]. However,
GAMT isolated from pig or rat liver is not inhibited by
creatine but is competitively inhibited by SAH, as well as the
other methyltransferases [27, 28]. Moreover, GAMT should
maintain the low levels of GAA, since it has been
demonstrated that high GAA levels result in neurotoxicity
[29]. Creatine supplementation is also known to modulate
methylation demand and decrease the plasma homocysteine
concentration [11, 30], brought about by the downregulation
of renal AGAT enzyme activity. Deminici et al. have shown
that creatine supplementation also prevents the decrease in
liver SAM concentration seen in high fat-fed rats [1].
2. HEPATIC INJURY AND CREATINE
Liver is a central organ involved in the regulation of
multimetabolic functions as well as detoxification of
xenobiotic, drugs, chemicals and infectious organism [31].
Therefore, several epidemiological studies have been
conducted to elucidate the main causes and consequences of
hepatic injury generation [32].
The development of liver injury was related to various
risk factors such as age, sex, alcohol, nutritional status, viral
infections, genetic factors, as well as the association of toxic
chemicals, fatty liver, nonalcoholic steatohepatitis (NASH),
drug induced liver injury (DILI), cirrhosis and liver cancer,
which could compromise the whole organism functionality
[33]. Among the myriad of metabolic problems resulting
from hepatic injury, the creatine endogenous synthesis is
also compromised [34]. In addition, a profound depletion of
brain creatine is intimately associated with severe
neurological symptoms that include mental retardation,
speech delay, and epileptic seizures [35].
Moreover, creatine exerts many of its functions by
increasing phosphocreatine levels, thereby permitting very
rapid regeneration of ATP after a burst of ATP utilization,
i.e. exercise conditions [35], and developing a spectrum of
therapeutic effects in both animals models and human
diseases [3, 36].
2.1. Possible Toxic Effects of Creatine Supplementation
in the Liver
After conducting studies that corroborated creatine
supplementation with ergogenic effects, this product has
become widely used for athletes and bodybuilders to
increase physical performance [37]. However, there are some
controversial studies about the ideal creatine dose which can
produce benefits without side effects [38, 39].
Research has revealed that creatine supplementation is
related to a reduction in the endogenous creatine formation,
down-regulation of renal AGAT activity [11], and
impairment of homocysteine (Hcy) production [30, 40].
Addition ally, Hcy reduction has been considered as one
mechanism of creatine neurological beneficial properties,
given the fact that high levels of Hcy increase the risk of
hypertension, neural tube defects, Alzheimer’s disease,
dementia, loss of cognitive function, and renal and liver
disease [7]. Thus, several researches showed that creatine
supplementation at “recommended” doses did not produce
toxic effects in the liver [41]. However, other authors have
reported that creatine supplementation leads to hepatocyte
injury as well as significant hepatic inflammatory lesions due
to direct toxic stress or intracellular creatine accumulation
[42]. Additionally, creatine accumulation may contribute to
the formation of cytotoxic substances, i.e. formaldehyde and
methylamine, due to the lower metabolic hepatocyte capacity
to convert creatine into creatinine and the enzymatic
capability of accomplishing methylation processes [43-45].
2.2. Uses of Creatine in Fatty Liver and NASH
NASH is a clinical patholog ical state of patients with
absence of alcohol abuse characterized by a wide spectrum
of liver damage including steatosis, non-alcoholic
steatohepatitis, fibrosis and cirrhosis [46, 47]. In addition, fat
accumulation and NASH progression are intimately
associated with the insufficiency of methionine metabolism
in liver, presenting a diminished availability of SAM as well
as an elevation in Hcy levels and oxidative stress generation
[1]. Moreover, normally the creatine biosynthesis uses SAM
stores and also produces Hcy in liver [6]. In this sense,
creatine supplementation is known to decrease the
consumption of SAM and also reduce the Hcy production in
liver, diminishing fat accu mulation in liver [1]. Recently, it
Creatine and Liver Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 15
has been indicated that creatine, but not structural analogs
such as guanidinoacetate, guanidinopropionic acid or or
methylguanidine, is able to reduce liv er triglycerides (TG)
synthesis and accumulation, thus increasing fatty acid
oxidation and TG secretion. These changes appear to be at
least partially mediated by the peroxisome proliferator of
activated receptor alpha (PPAR-α) pathway signaling and its
downstream targets [48].
2.3. Disorders by Creatine Supplementation and its
Utility in Acute Liver Failure Associated with ELA
Encephalopathy
Creatine, phosphocreatine and creatine kinase are
important in maintaining cellular energy homeostasis that
would prevent or impede the neurodegenerative process [49,
50]. Thus, creatine supplementation has been used in a
number of neurological and neurodegenerative diseases such
as Parkinson’s disease, Huntington’s disease, amyotrophic
lateral sclerosis, and Alzheimer’s disease [51-53]. Moreover,
several studies have associated different drugs, i.e.
acetaminophen, thioacetamide or carbon tetrachloride, with
acute liver failure and encephalopathy development [54, 55].
This encephalopathy is intimately linked with increased
ammonia levels in blood and brain, which can diminish the
energy in brain and decrease neuronal activity as well [56].
Different studies have shown that creatine supplementation
could supply brain energy, presenting neuroprotective effects
against the encephalopathy induced by hyperammonemia
[49, 56]. Curiously, the lack of energy caused by
hyperammonemia, which is involved with neuronal death,
seems to b e able to increase the creatin e uptake in brain
endothelial cells [49, 56, 57].
3. FINAL CONSIDERATIONS
Creatine, in the form of creatine monohydrate, is taken
by many athletes for its ergogenic properties [35] and
performance-enhancing potential [58-62], and some human
studies have demonstrated benefits in certain pathological
conditions [63-66]. On the other hand, little is known about
the adverse effects of creatine supplementation, which are
barely described in the literature [67, 68], with reports of
mainly hypothetical effects arising from a small number of
scientific publications.
In animals, the effects of creatine supplementation on
liver have not been well described and some of them are
conflicting. Whereas some studies did not observe any
alteration in renal and hepatic function [42, 69], others have
reported that creatine supplementation can accelerate renal
and hepatic disease progression [42, 70-72]. The potential
creatine toxicity is based on some anecdotal human case
reports [73, 74], one animal study in hypertensive rats [70],
and the fact that carcinogens can be formed if creatine and
sugars are heated to high temperatures [16, 75].
In humans, the most commonly recommended dose of
creatine supplementation is 5g/day [41, 58, 61], however, it
can range from 3 g/day to 20 g/day [59, 60, 62, 68].
Moreover, most of the studies that have examined the
potential for toxicity of creatine supplementation in humans,
have not found evidence of side effects when consumed at
recommended doses [41, 62, 76-81]. Little information on
liver metabolism changes induced by oral creatine
supplementation is available. There are studies reporting
some data on liver function while consuming creatine
supplements [82-84]. The results of these studies indicate no
or minimal changes on the level of serum urea and bilirubin,
alkaline phosphatase, aspartate aminotransferase, alanine
aminotransferase and ɣ -glutamyltransferase. The general
conclusion is that creatine supplementation induces no real
modification in liver metabolism in humans.
However, some isolated cases have been found in the
literature. For example, a case report identified a healthy
individual that had been using creatine supplementation
during 9 months accompanied by a supplementation of whey
protein on the last four months. The blood analysis of this
individual demonstrated high level of aminotransferases,
alkaline phosphate, bilirubin and creatinine. Symptoms
decreased gradually after the supplementation withdrawn.
The liver analysis showed findings similar to a drug-induced
cholestasis [85].
The possible side effects of creatine supplementation,
regarding liver and renal toxicity, or dysfunction are still
inconclusive [86]. It is speculated that creatine
supplementation can interfere on urea metabolism, which is
one of the metabolic products of creatine metabolism. Urea
is involved in the conversion of toxic compounds such as
methylamine and formaldehyde, and creatine supplementation
can also be expected to influence this conversion [44].
Another curious data found in literature is that creatine
supplementation decreased blood glucose level in mice,
possibly due to the fact that it leads to increased insulin
production [87]. This study supported data by Rooney et al.,
who reported that chronic supplementation of creatine leads
to higher secretion of insulin, which decreases the blood
glucose concentrations [88].
There also some contradictory theories regarding the
oxidant/antioxidant effect of creatine. Antioxidant effects
were found in several studies with creatine supplementation
[89], decreasing markers of oxidative stress in animal
models of neurodegenerative disease [90, 91]. In liver of
exercise rats, it has been reported that creatine
supplementation could improve the activity of some
antioxidant enzymes such as glutathione peroxidase and
catalase, but it was not effective in normalizing the increased
hydrogen peroxide (H2O2) concentrations induced by
exercise in rats [92]. Interestingly, other studies indicated
that creatine can produce antioxidant effects on liver
resistance-exercised rats, scavenging reactive oxygen species
but with no changes in th e activity of antioxidants enzymes
[93]. On the other way, one of the theories regarding the
potential for toxicity from creatine supplementation is that
creatine can increase oxidative stress and potentially form
carcinogenic compounds in vitro [16]. Tarnopolsky et al.
have shown that creatine supplementation in regular doses
results on a lymphocytic hepatitis in a mouse model of
oxidative stress [42]. It was postulated that either direct toxic
stress or osmotic changes after 40 days of creatine
supplementation in rats submitted to swimming training
result on increases in AST and GGT levels. This study,
suggests that creatine can promote substantial changes in
16 Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 Barcelos et al.
liver and kidney metabolism and/or function, possibly
developing hepatic and renal disorders [94]. Moreover, other
studies confirmed that long-term creatine supplementation
(4-8 weeks) can also induce structural alterations, indicating
hepatic and renal damage, in sedentary rats [95].
However, in spite of the above commented cases, the
general conclusion about creatine supplementation toxicity is
the fact that the major risk for health is probably associated
with the purity of commercially available creatine [96].
Nevertheless, there is no conclusive evidence to affirm that
this supplementation may affect kidney and liver function in
healthy individuals [62, 76-81, 97-102].
LIST OF ABBREVIATIONS
AGAT = L-arginine:glycine amidinotransferase
DILI = Drug induced liver injury
GAA = Guanidinoacetic acid
GAMT = Guanidinoacetate N-methyltransferase
Hcy = Homocysteine
NASH = Nonalcoholic steatoshepatitis
PPARα = Peroxisome proliferator of activated receptor
alpha
SAH = S-adenosylhomocysteine
SAM = S-adenosylmethionine
TG = Triglycerides
CONFLICT OF INTEREST
The author(s) confirm that this article content has no
conflict of interest.
ACKNOWLEDGEMENTS
CIBERehd is funded by the Instituto de Salud Carlos III,
Spain. Coordenação de Aperfeiçoamento de Pessoal de Nível
Superior (CAPES) for providing fellowship to R.P.B. and
S.T.S. Conselho Nacional de Desenvolvimento Científico e
Tecnológico (CNPq) for providing fellowship to F.A.A.S.
REFERENCES
[1] Deminice, R.; da Silva, R. P.; Lamarre, S. G.; Brown, C.; Furey, G.
N.; McCarter, S. A.; Jordao, A. A.; Kelly, K. B.; King-Jones, K.;
Jacobs, R.L.; Brosnan, M.E.; Brosnan, J.T. Creatine supplementation
prevents the accumulation of fat in the livers of rats fed a high-fat
diet. J. Nutr., 2011, 141(10), 1799-1804.
[2] da Silva, R. P.; Nissim, I.; Brosnan, M. E.; Brosnan, J. T. Creatine
synthesis: hepatic metabolism of guanidinoacetate and creatine in
the rat in vitro and in vivo. Am. J. Physiol. Endocrinol. Metab.,
2009, 296(2), E256-261.
[3] Persky, A. M.; Brazeau, G. A. Clinical pharmacology of the dietary
supplement creatine monohydrate. Pharmacol. Rev., 2001, 53, 161-
176.
[4] Aoyagi, K.; Akiyama, K.; Kuzure, Y.; Takemura, K.; Nagase, S.;
Ienaga, K.; Nakamura, K.; Koyama, A.; Narita, M. Synthesis of
creatol, a hydroxyl radical adduct of creatinine and its increase by
puromycin aminonucleoside in isolated rat hepatocytes. Free
Radic. Res., 1998, 29(3), 221-226.
[5] Walker, J. B. Creatine: biosynthesis, regulation, and function. Adv.
Enzymol. Rela t. Areas Mol. Biol., 1979, 50, 177-242.
[6] Stead, L. M.; Brosnan, J. T.; Brosnan, M. E.; Vance, D. E.; Jacobs,
R. L. Is it time to reevaluate methyl balance in humans? Am. J.
Clin. Nutr., 2006, 83(1), 5-10.
[7] Wyss, M.; Schulze, A. Health implications of creatine: can oral
creatine supplementation protect against neurological and
atherosclerotic disease? Neuroscience, 2002, 112(2), 243-260.
[8] Askanas, V.; McFerrin, J.; Baque, S.; Alvarez, R. B.; Sarkozi, E.;
Engel, W. K. Transfer of beta-amyloid precursor protein gene using
adenovirus vector causes mitochondrial abnormalities in cultured
normal human muscle. Proc. Natl. Acad. Sci. U.S.A., 1996, 93(3),
1314-1319.
[9] Guthmiller, P.; Van Pilsum, J. F.; Boen, J. R.; McGuire, D. M.
Cloning and sequencing of rat kidney Larginine:glycine
amidinotransferase. Studies on the mechanism of regulation by
growth hormone and creatine. J. Biol. Chem., 1994, 269, 17556-
17560.
[10] Stockler, S.; Marescau, B.; De Deyn, P. P.; Trijbels, J. M. ;
Hanefeld, F. Guanidino compounds in guanidinoacetate
methyltransferase deficiency, a new inborn error of creatine
synthesis. Metabolism, 1997, 46(10), 1189-1193.
[11] Edison, E. E.; Brosnan, M. E.; Meyer, C.; Brosnan, J. T. Creatine
synthesis: production of guanidinoacetate by the rat and human
kidney in vivo. Am. J. Physiol. Renal Physiol., 2007, 293(6),
F1799-1804.
[12] Fitch, C. D.; Hsu, C.; Dinning, J. S. The mechanism of kidney
transamidinase reduction in vitamin Edeficient rabbits. J. Biol.
Chem., 1961, 236, 490-492.
[13] Goldman, R.; Moss, J. X. Creatine synthesis after creatinine
loading and after nephrectomy. Proc. Soc. Exp. Biol. Med., 1960,
105, 450-453.
[14] Levillain, O.; Marescau, B.; de Deyn, P. P. Gu anidino co mpound
metabolism in rats subjected to 20% to 90% nephrectomy. Kidney
Int., 1995, 47(2), 464-472.
[15] Adams, G. R.; Baldwin, K. M. Age dependence of myosin heavy
chain transitions induced by creatine depletion in rat skeletal
muscle. J. Appl. Physiol., 1995, 78(1), 368-371.
[16] Wyss, M.; Kaddurah-Daouk, R. Creatine and creatinine
metabolism. Physiol. Rev., 2000, 80(3), 1107-1213. [17] Aired, S.;
Creach, Y.; Palevody, C.; Esclassan, J.; Hollande, E. Creatine
phosphate as energy source in the cerulein-stimulated rat pancreas
study by 31P nuclear magnetic resonance. Int. J. Pancreatol., 1991,
10(1), 81-95.
[17] Askenasy, N.; Koretsky, A. P. Differential effects of creatine
kinase isoenzymes and substrates on regeneration in livers of
transgenic mice. Am. J. Physiol., 1997, 273(2 Pt1), C741-C746.
[18] Alsever, R. N.; Georg, R. H.; Sussman, K. E. Stimulation of insulin
secretion by guanidinoacetic acid and other guanidine derivatives.
Endocrinology, 1970, 86, 332-336.
[19] Ambrosio, G.; Zweier, J.L.; Flaherty, J.T. The relationship between
oxygen radical generation and impairment of myocardial energy
metabolism following post-ischemic reperfusion. J. Mol. Cell.
Cardiol., 1991, 23(12), 1359-1374.
[20] Adams, G. R.; Bodell, P. W.; Baldwin, K. M. Running
performance and cardiovascular capacity are not impaired in
creatine-depleted rats. J. Appl. Physiol., 1995, 79(3), 1002-1007.
[21] Speer, O.; Neukomm, L. J.; Murphy, R. M.; Zanolla, E.; Schlattner,
U.; Henry, H.; Snow, R. J.; Wallimann, T. Creatine transporters: a
reappraisal. Mol. Cell. Biochem., 2004, 256-257(1-2), 407-424.
[22] Alink, G. M.; Knize, M. G.; Shen, N. H.; Hesse, S. P.; Felton, J. S.
Mutagenicity of food pellets from human diets in The Netherlands.
Mutat. Res., 1988, 206(1), 387-393.
[23] Altschuld, R. A.; Gamelin, L. M.; Kelley, R. E.; Lambert, M. R.;
Apel, L. E.; Brierley, G. P. Degradation and resynthesis of adenine
nucleotides in adult rat heart myocytes. J. Biol. Chem., 1987,
262(28), 1352713533.
[24] McGuire, D. M.; Gross, M. D.; Van Pilsum, J. F.; Towle, H. C.
Repression of rat kidney L-arginine:glycine amidinotransferase
synthesis by creatine at a pretranslational level. J. Biol. Chem.,
1984, 259(19), 1203412038.
[25] Aksenova, M. V.; Aksenov, M. Y.; Payne, R. M.; Trojanowski, J.
Q.; Schmidt, M. L.; Carney, J. M.; Butterfield, D. A.; Markesbery,
W. R. Oxidation of cytosolic proteins and expression of creatine
kinase BB in frontal lobe in different neurodegenerative disorders.
Dement. Geriatr. Cogn. Disord., 1999, 10(2), 158165.
Creatine and Liver Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 17
[26] Akamatsu, S.; Miyashita, R. Bacterial decomposition of creatine.
III. The pathway of creatine decomposition. Enzymologia, 1952,
15(4), 173-176.
[27] Aliev, M. K.; van Dorsten, F. A.; Nederhoff, M. G.; van Echteld,
C. J.; Veksler, V.; Nicolay, K.; Saks, V.A. Mathematical model of
compartmentalized energy transfer: its use for analysis and
interpretation of 31PNMR studies of isolated heart of creatine
kinase deficient mice. Mol. Cell. Biochem., 1998, 184(3), 209229.
[28] Ambrosio, G.; Jacobus, W. E.; Bergman, C. A.; Weisman, H. F.;
Becker, L. C. Preserved high energy phosphate metabolic reserve
in globally "stunned" hearts despite reduction of basal ATP content
and contractility. J. Mol. Cell. Cardiol., 1987, 19(10), 953-964.
[29] Deminice, R.; Portari, G. V.; Vannucchi, H.; Jordao, A. A. Effects
of creatine supplementation on homocysteine levels and lip id
peroxidation in rats. Br. J. Nutr., 2009, 102(1), 110-116.
[30] Ingawale, D. K.; Mandlik, S. K.; Naik, S. R. Models of
hepatotoxicity and the underlying cellular, biochemical and
immunological mechanism(s): a critical discussion. Environ.
Toxicol. Pharmacol., 2014, 37(1), 118-133.
[31] Jalan, R.; Gines, P.; Olson, J. C.; Mookerjee, R. P.; Moreau, R.;
Garcia-Tsao, G.; Arroyo, V.; Kamath, P. S. Acute-on chronic liver
failure. J. Hepatol., 2012, 57(6), 1336-1348.
[32] Wang, K. Molecular mechanisms of hepatic apoptosis. Cell Death
Dis., 2014, 5, e996.
[33] Kharbanda, K. K.; Todero, S. L.; Moats, J. C.; Harris, R. M.; Osna,
N. A.; Thomes, P. G.; Tuma, D. J. Alcohol consumption decreases
rat hepatic creatine biosynthesis via altered guanidinoacetate
methyltransferase activity. Alcohol. Clin. Exp. Res., 2014, 38(3),
641-648.
[34] Brosnan, J. T.; Brosnan, M. E. Creatine: endogenous metabolite,
dietary, and therapeutic supplement. Annu. Rev. Nutr., 2007, 27,
241-261.
[35] Klein, A.; Ferrante, R. In:Creatine and creatine kinase in health and
disease; Salomons, G., Wyss, M., Eds.; Springer Netherlands:
2007, 46, pp. 205-243.
[36] Maughan, R. J.; King, D. S.; Lea, T. Dietary supplements. J. Sports
Sci., 2004, 22(1), 95-113.
[37] Brudnak, M. A. Creatine: are the benefits worth the risk? Toxicol.
Lett., 2004, 150(1), 123-130.
[38] Wyss, M. Writing about creatine: is it worth the risk? Toxicol. Lett.,
2004, 152(3), 273-274.
[39] Stead, L. M.; Au, K. P.; Jacobs, R. L.; Brosnan, M. E.; Brosnan, J.
T. Methylation demand and homocysteine metabolism: effects of
dietary provision of creatine and guanidinoacetate. Am. J. Physiol.
Endocrinol. Metab., 2001, 281(5), E1095-1000.
[40] Kreider, R. B.; Melton, C.; Rasmussen, C. J.; Greenwood, M.;
Lancaster, S.; Cantler, E. C.; Milnor, P.; Almada, A. L. Long-term
creatine supplementation does not significantly affect clinical
markers of health in athletes. Mol. Cell. Biochem., 2003, 244(1-2),
95-104.
[41] Tarnopolsky, M. A.; Bourgeois, J. M.; Snow, R.; Keys, S.; Roy, B.
D.; Kwiecien, J. M.; Turnbull, J. Histological assessment of
intermediate- and long-term creatine monohydrate supplementation
in mice and rats. Am. J. Physiol. Regul. Integr. Comp. Physiol.,
2003, 285(1), R762-769.
[42] Clayton, T. A.; Lindon, J. C.; Everett, J. R.; Charuel, C.; Hanton,
G.; Le Net, J. L.; Provost, J. P.; Nicholson, J. K. Hepatotoxin-
induced hypercreatinaemia and hypercreatinuria: their relationship
to one another, to liver damage and to weakened nutritional status.
Arch. Toxicol., 2004, 78(2), 86-96.
[43] Poortmans, J. R.; Kumps, A.; Duez, P.; Fofonka, A.; Carpentier,
A.; Francaux, M. Effect of oral creatine supplementation on urinary
methylamine, formaldehyde, and formate. Med. Sci. Sports Exerc.,
2005, 37(10), 1717-1720.
[44] Yu, P.H.; Deng, Y. Potential cytotoxic effect of chronic
administration of creatine, a nutrition supplement to augment
athletic performance. Med. Hypotheses, 2000, 54(5), 726-728.
[45] Lima-Cabello, E.; Garcia-Mediavilla, M. V.; Miquilena-Colina, M.
E.; Vargas-Castrillon, J.; Lozano-Rodriguez, T.; Fernandez-
Bermejo, M.; Olcoz, J. L.; Gonzalez-Gallego, J.; Garcia-Monzon,
C.; SanchezCampos, S. Enhanced expression of pro-inflammatory
mediators and liver X-receptor-regulated lipogenic genes in non-
alcoholic fatty liver disease and hepatitis C. Clin. Sci. (Lond), 2011,
120(6), 239-250.
[46] Ordoñez, R.; Carbajo-Pescador, S.; Mauriz, J. L.; González-
Gallego, J. Understanding nutritional interventions and physical
exercise in non-alcoholic fatty liver disease. Curr. Mol. Med.,
2015, 15(1), 3-6.
[47] da Silva, R. P.; Kelly, K. B .; Leonard, K. A.; Jacobs, R. L. Creatine
reduces hepatic TG accumulation in hepatocytes by stimulating
fatty acid oxidation. Biochim. Biophys. Acta, 2014, 1841(11), 1639-
1646.
[48] Belanger, M.; Asashima, T.; Ohtsuki, S.; Yamaguchi, H.; Ito, S.;
Terasaki, T. Hyperammonemia induces transport of taurine and
creatine and suppresses claudin-12 gene expression in brain
capillary endothelial cells in vitro. Neurochem. Int., 2007, 50(1),
95-101.
[49] Wallimann, T.; Wyss, M.; Brdiczka, D.; Nicolay, K.; Eppenberger,
H. M. Intracellular compartmentation, structure and function of
creatine kinase isoenzymes in tissues with high an d fluctuating
energy demands: the 'phosphocreatine circuit' for cellular energy
homeostasis. Biochem. J., 1992, 281(Pt 1), 21-40.
[50] Andres, R. H.; Ducray, A. D.; Schlattner, U.; Wallimann, T.;
Widmer, H. R. Functions and effects of creatine in the central
nervous system. Brain Res. Bul l., 2008, 76(4), 329-343.
[51] Andreassen, O. A.; Jenkins, B. G.; Dedeoglu, A.; Ferrante, K. L.;
Bogdanov, M. B.; Kaddurah-Daouk, R.; Beal, M. F. Increases in
cortical glutamate concentrations in transgenic amyotrophic lateral
sclerosis mice are attenuated by creatine supplementation. J.
Neurochem., 2001, 77(2), 383-390.
[52] Groeneveld, G. J.; Veldink, J. H.; van der Tweel, I.; Kalmijn, S.;
Beijer, C.; de Visser, M.; Wokke, J. H.; Franssen, H.; van den
Berg, L. H. A randomized sequential trial of creatine in
amyotrophic lateral sclerosis. Ann. Neurol., 2003, 53(4), 437-445.
[53] Butterworth, R. F.; Norenberg, M. D.; Felipo, V.; Ferenci, P.;
Albrecht, J.; Blei, A. T.; Members of the ISHEN Commission on
Experimental Models of HE Experimental models of hepatic
encephalopathy: ISHEN guidelines. Liver Int., 2009, 29(6), 783-
788.
[54] Mullen, K.D.; Birgisson, S.; Gacad, R.C.; Conjeevaram, H. Animal
models of hepatic encephalopathy and hyperammonemia. Adv. Exp.
Med. Biol., 1994, 368, 1-10.
[55] Skowronska, M.; Albrecht, J. Alterations of blood brain barrier
function in hyperammonemia: an overview. Neurotox Res., 2012,
21(2), 236-244.
[56] Braissant, O.; Henry, H.; Villard, A. M.; Zurich, M. G.; Loup, M.;
Eilers, B.; Parlascino, G.; Matter, E.; Boulat, O.; Honegger, P.;
Bachmann, C. Ammonium-induced impairment of axonal growth is
prevented through glial creatine. J. Neurosci., 2002, 22(22), 9810-
9820.
[57] Greenhaff, P. L.; Casey, A.; Short, A. H.; Harris, R.; Soderlund, K.;
Hultman, E. Influence of oral creatine supplementation of muscle
torque during repeated bouts of maximal voluntary exercise in man.
Clin. Sci. (Lond), 1993, 84(5), 565-571.
[58] Grindstaff, P. D.; Kreider, R.; Bishop, R.; Wilson, M.; Wood, L.;
Alexander, C.; Almada, A. Effects of creatine supplementation on
repetitive sprint performance and body composition in competitive
swimmers. Int. J. Sport Nutr., 1997, 7(4), 330-346.
[59] Maganaris, C. N.; Maughan, R. J. Creatine supplementation
enhances maximum voluntary isometric force and endurance
capacity in resistance trained men. Acta Physiol. Scand., 1998,
163(3), 279-287.
[60] Tarnopolsky, M. A.; MacLennan, D. P. Creatine monohydrate
supplementation enhances high-intensity exercise performance in
males and females. Int. J. Sport Nutr. Exerc. Metab., 2000, 10(4),
452-463.
[61] Terjung, R. L.; Clarkson, P.; Eichner, E. R.; Greenhaff, P. L.;
Hespel, P. J.; Israel, R. G.; Kraemer, W. J.; Meyer, R. A.; Spriet, L.
L.; Tarnopolsky, M. A.; Wagenmakers, A. J.; Williams, M. H.
American College of Sports Medicine roundtable. The
physiological and health effects of oral creatine supplementation.
Med. Sci. Sports Exerc., 2000, 32(3), 706-717.
[62] Leuzzi, V.; Bianchi, M. C.; Tosetti, M.; Carducci, C.; Cerquiglini,
C. A.; Cioni, G.; Antonozzi, I. Brain creatine depletion:
guanidinoacetate methyltransferase deficiency (improving with
creatine supplementation). Neurology, 2000, 55(9), 1407-1409.
[63] Tarnopolsky, M.; Martin, J. Creatine monohydrate increases
strength in patients with neuromuscular disease. Neurology, 1999,
52(4), 854-857.
18 Mini-Reviews in Medicinal Chemistry, 2016, Vol. 16, No. 1 Barcelos et al.
[64] Tarnopolsky, M.A.; Roy, B.D.; MacDonald, J.R. A randomized,
controlled trial of creatine monohydrate in patients with
mitochondrial cytopathies. Muscle Nerve, 1997, 20(12), 1502-
1509.
[65] Walter, M.C.; Lochmuller, H.; Reilich, P.; Klopstock, T.; Huber,
R.; Hartard, M.; Hennig, M.; Pongratz, D.; Muller-Felber, W.
Creatine monohydrate in muscular dystrophies: A double-blind,
placebo-controlled clinical study. Neurology, 2000, 54(9), 1848-
1850.
[66] Juhn, M. S.; Tarnopolsky, M. Potential side effects of oral creatine
supplementation: a critical review. Clin. J. Sport Med., 1998, 8(4),
298-304.
[67] Demant, T. W.; Rhodes, E. C. Effects of creatine supplementation
on exercise performance. Sports Med., 1999, 28(1), 49-60.
[68] Taes, Y. E.; Delanghe, J. R.; Wuyts, B.; van de Voorde, J.;
Lameire, N. H. Creatine supplementation does not affect kidney
function in an animal model with pre-existing renal failure.
Nephrol. Dial. Transplant., 2003, 18(2), 258-264.
[69] Edmunds, J. W.; Jayapalan, S.; DiMarco, N. M.; Saboorian, M. H.;
Aukema, H. M. Creatine supplementation increases renal disease
progression in Han:SPRD-cy rats. Am. J. Kidney Dis., 2001, 37(1),
73-78.
[70] Ferreira, L. G.; De Toledo Bergamaschi, C.; Lazaretti-Castro, M.;
Heilberg, I. P. Effects of creatine supplementation on body
composition and renal function in rats. Med. Sci. Sports Exerc.,
2005, 37(9), 15251529.
[71] Thorsteinsdottir, B.; Grande, J. P.; Garovic, V. D. Acute renal
failure in a young weight lifter taking multiple food supplements,
including creatine monohydrate. J. Ren. Nutr., 2006, 16(4), 341-
345.
[72] Koshy, K. M.; Gr iswold, E.; Schneeberger, E. E. Interstitial
nephritis in a patient taking creatine. N. Engl. J. Med., 1999,
340(10), 814-815.
[73] Pritchard, N.R.; Kalra, P.A. Renal dysfunction accompanying oral
creatine supplements. Lancet, 1998, 351(9111), 1252-1253.
[74] Yoshida, D.; Okamoto, H. Formation of mutagens by heating
creatine and glucose. Biochem. Biophys. Res. Commun., 1980,
96(2), 844-847.
[75] Mayhew, D. L.; Mayhew, J. L.; Ware, J. S. Effects of long-term
creatine supplementation on liver and kidney functions in
American college football players. Int. J. Sport Nutr. Exerc.
Metab., 2002, 12(4), 453460.
[76] Mihic, S.; MacDonald, J. R.; McKenzie, S.; Tarnopolsky, M. A.
Acute creatine loading increases fat-free mass, but does not affect
blood pressure, plasma creatinine, or CK activity in men and
women. Med. Sci. Sports Exerc., 2000, 32(2), 291-296.
[77] Poortmans, J. R.; Francaux, M. Long-term oral creatine
supplementation does not impair renal function in healthy athletes.
Med. Sci. Sports Exerc., 1999, 31(8), 1108-1110.
[78] Poortmans, J. R.; Auquier, H.; Renaut, V.; Durussel, A.; Saugy,
M.; Brisson, G. R. Effect of short-term creatine supplementation on
renal responses in men. Eur. J. Appl. Physiol. Occup. Physiol.,
1997, 76(6), 566-567.
[79] Robinson, T. M.; Sewell, D. A.; Casey, A.; Steenge, G.; Greenhaff,
P. L. Dietary creatine supplementation does not affect some
haematological indices, or indices of muscle damage and hepatic
and renal function. Br. J. Sports Med., 2000, 34(4), 284-288.
[80] Waldron, J. E.; Pendlay, G. W.; Kilgore, T. G.; Haff, G. G.; Reeve,
J. S. Concurrent creatine monohydrate supplementation and
resistance training does not affect markers of hepatic function in
trained weightlifters. JEP Online, 2002, 5(1), 57-64.
[81] Earnest, C.; Almada, A.; Mitchell, T. Influence of chronic creatine
supplementation on hepatorenal function. FASEB J., 1996, 10,
A790.
[82] Almada, A.; Mitchell, T.; Earnest, C. Impact of chronic creatine
supplementation on serum enzyme concentration. FASEB J., 1996,
10, A791.
[83] Mihic, S.; MacDonald, J. R.; McKenzie, S.; Tarnopolsky, M. A.
The effect of creatine supplementation on blood pressure, plasma
creatine kinase, and body composition. FASEB J., 1998, 12, A652.
[84] Whitt, K. N.; Ward, S. C.; Deniz, K.; Liu, L. ; Odin, J. A.; Qin, L.
Cholestatic liver injury associated with whey protein and creatine
supplements. Semin. Liver Dis., 2008, 28(2), 226-231.
[85] Bizzarini, E.; De Angelis, L. Is the use of oral creatine
supplementation safe? J. Sports Med. Phys. Fitness, 2004, 44(4),
411-416.
[86] Iqbal, S.; Nazir, N.; Gillani, Q.; Akbar, A.; Iqbal, F . Effect of
creatine monohydrate supplementation on various hematological
and serum biochemical parameters of male albino mice following
neonatal hypoxiaischemia encephalopathy. Sci. World J., 2013,
2013, 286075.
[87] Rooney, K.; Bryson, J.; Phuyal, J.; Denyer, G.; Caterson, I.;
Thompson, C. Creatine supplementation alters insulin secretion and
glucose homeostasis in vivo. Metabolism, 2002, 51(4), 518-522.
[88] Lawler, J. M.; Barnes, W. S.; Wu, G.; Song, W.; Demaree, S.
Direct antioxidant properties of creatine. Biochem. Biophys. Res.
Commun., 2002, 290(1), 47-52.
[89] Klivenyi, P.; Ferrante, R. J.; Matthews, R. T.; Bogdanov, M. B.;
Klein, A. M.; Andreassen, O. A.; Mueller, G.; Wermer, M.;
Kaddurah-Daouk, R.; Beal, M. F. Neuroprotective effects of
creatine in a transgenic animal model of amyotrophic lateral
sclerosis. Nat. Med., 1999, 5(3), 347-350.
[90] Matthews, R. T.; Yang, L.; Jenkins, B. G.; Ferrante, R. J.; Rosen,
B. R.; Kaddurah-Daouk, R.; Beal, M. F. Neuroprotective effects of
creatine and cyclocreatine in animal models of Huntington's
disease. J. Neurosci., 1998, 18(1), 156-163.
[91] Araujo, M. B.; Moura, L. P.; Junior, R. C.; Junior, M. C.; Dalia, R.
A.; Sponton, A. C.; Ribeiro, C.; Mello, M. A. Creatine
supplementation and oxidative stress in rat liver. J. Int. Soc. Sports
Nutr., 2013, 10(1), 542783-10-54.
[92] Stefani, G. P.; Nunes, R. B.; Dornelles, A. Z.; Alves, J. P.; Piva, M.
O.; Domenico, M. D.; Rhoden, C. R.; Lago, P. D. Effects of
creatine supplementation associated with resistance training on
oxidative stress in different tissues of rats. J. Int. Soc. Sports Nutr.,
2014, 11(1), 11-2783-11-11.
[93] Souza, W. M.; Heck, T. G.; Wronski, E. C.; Ulbrich, A. Z.; Boff, E.
Effects of creatine supplementation on biomarkers of hepatic and
renal function in young trained rats. Toxicol. Mech. Methods, 2013,
23(9), 697701.
[94] Souza, R. A.; Miranda, H.; Xavier, M.; Lazo-Osorio, R. A.;
Gouvea, H. A.; Cogo, J. C.; Vieira, R. P.; Ribeiro, W. Effects of
high-dose creatine supplementation on kidney and liver responses
in sedentary and exercised rats. J. Sports Sci. Med., 2009, 8(4),
672-681.
[95] Francaux, M.; Poortmans, J. R. Side effects of creatine
supplementation in athletes. Int. J. Sports Physiol. Perform., 2006,
1(4), 311-323.
[96] Farquhar, W. B.; Zambraski, E. J. Effects of creatine use on the
athlete's kidney. Curr. Sports Med. Rep., 2002, 1(2), 103-106.
[97] Groeneveld, G. J.; Beijer, C.; Veldink, J. H.; Kalmijn, S.; Wokke,
J. H.; van den Berg, L. H. Few adverse effects of long-term
creatine supplementation in a placebo-controlled trial. Int. J. Sports
Med., 2005, 26(4), 307-313.
[98] Kreider, R. B. Effects of creatine supplementation on performance
and training adaptations. Mol. Cell. Biochem., 2003, 244(1-2), 89-
94.
[99] Pline, K. A.; Smith, C. L. The effect of creatine intake on renal
function. Ann. Pharmacother., 2005, 39(6), 1093-1096.
[100] Poortmans, J.R.; Francaux, M. Adverse effects of creatine
supplementation: fact or fiction? Sports Med., 2000, 30(3), 155-170.
[101] Yoshizumi, W.M.; Tsourounis, C. Effects of creatine supplementation
on renal function. J. Herb Pharmacother., 2004, 4(1), 1-7.
Received: January 20, 2015 Revised: March 21, 2015 Accepted: April 23, 2015
... These processes involve the following: l-arginine glycine aminotransferase (AGAT) converts l-arginine and glycine into guanidinoacetate (GAA) and l-ornithine in the mitochondrial intermembrane space; N-guanidinoacetate methyltransferase (GAMT) transfers a methyl group from S-adenosylmethionine (SAM) to GAA to produce creatine; and a specific creatine plasma membrane transporter, SLC6A8, is observed in the kidney, brain, and liver. 16,17 . Dietary Cr enters the brain through the bloodbrain barrier (BBB) through SLC6A8, which is expressed in the microcapillaries of the BBB, neurons, and oligodendrocytes but not in perivascular astrocytes [17][18][19] . ...
... GAMT expression is low in PC cells, and might act as a tumor suppressor [20,21]. As an essential enzyme in the creatine-biosynthetic pathway [22], GAMT can convert the glycine metabolite guanidoacetate to creatine, which mediates AMPK activation [23,24]. AMPK can induce caspase-family members, leading to cancer cell apoptosis [25,26], although the corresponding mechanism of GAMT in PC cell apoptosis remains unclear. ...
Article
Full-text available
Background Pancreatic cancer (PC) is an extremely malignant tumor with low survival rate. Effective biomarkers and therapeutic targets for PC are lacking. The roles of circular RNAs (circRNAs) in cancers have been explored in various studies, however more work is needed to understand the functional roles of specific circRNAs. In this study, we explore the specific role and mechanism of circ_0035435 (termed circCGNL1) in PC. Methods qRT-PCR analysis was performed to detect circCGNL1 expression, indicating circCGNL1 had low expression in PC cells and tissues. The function of circCGNL1 in PC progression was examined both in vitro and in vivo. circCGNL1-interacting proteins were identified by performing RNA pulldown, co-immunoprecipitation, GST-pulldown, and dual-luciferase reporter assays. Results Overexpressing circCGNL1 inhibited PC proliferation via promoting apoptosis. CircCGNL1 interacted with phosphatase nudix hydrolase 4 (NUDT4) to promote histone deacetylase 4 (HDAC4) dephosphorylation and subsequent HDAC4 nuclear translocation. Intranuclear HDAC4 mediated RUNX Family Transcription Factor 2 (RUNX2) deacetylation and thereby accelerating RUNX2 degradation. The transcription factor, RUNX2, inhibited guanidinoacetate N-methyltransferase (GAMT) expression. GAMT was further verified to induce PC cell apoptosis via AMPK–AKT–Bad signaling pathway. Conclusions We discovered that circCGNL1 can interact with NUDT4 to enhance NUDT4-dependent HDAC4 dephosphorylation, subsequently activating HDAC4–RUNX2–GAMT-mediated apoptosis to suppress PC cell growth. These findings suggest new therapeutic targets for PC.
... Lastly, those with high blood pressure or liver disease should not take creatine. Individuals who consume significant amounts of caffeine, take diuretics, or are on medications that affect kidney function should proceed with caution (Mount Sinai), (Barcelos et al., 2016;Bizzarini & De Angelis, 2004;Brewster et al., 2006;Poortmans & Francaux, 2000). It is recommended that an individual consult with their health care provider before supplementing creatine into their diets. ...
Article
Full-text available
Creatine is a popular and widely used ergogenic dietary supplement among athletes, for which studies have consistently shown increased lean muscle mass and exercise capacity when used with short-duration, high-intensity exercise. This article provides an overview of creatine supplementation, particularly in the context of athletes, focusing on its safety, benefits, dosage, and considerations for young individuals. Research has shown that creatine supplementation may provide additional benefits including enhanced post-exercise recovery, injury prevention, and rehabilitation, as well as several potential neurological benefits that may be relevant to sports. Studies show that short- and long-term supplementation is safe and well-tolerated in healthy individuals and several patient populations.
... These processes involve the following: l-arginine glycine aminotransferase (AGAT) converts l-arginine and glycine into guanidinoacetate (GAA) and l-ornithine in the mitochondrial intermembrane space; N-guanidinoacetate methyltransferase (GAMT) transfers a methyl group from S-adenosylmethionine (SAM) to GAA to produce creatine; and a specific creatine plasma membrane transporter, SLC6A8, is observed in the kidney, brain, and liver. 16,17 . Dietary Cr enters the brain through the bloodbrain barrier (BBB) through SLC6A8, which is expressed in the microcapillaries of the BBB, neurons, and oligodendrocytes but not in perivascular astrocytes [17][18][19] . ...
Article
[Purpose] The maintenance of energy balance in the body, especially in energy-demanding tissues like the muscles and the central nervous system, depends on creatine (Cr). In addition to improving muscle function, Cr is necessary for the bioenergetics of the central nervous system because it replenishes adenosine triphosphate without needing oxygen. Furthermore, Cr possesses anti-oxidant, anti-apoptotic, and anti-excitotoxic properties. Clinical research on neurodegenerative illnesses has shown that Cr supplementation results in less effective outcomes. With a brief update on the possible role of Cr in human, animal, and in vitro experiments, this review seeks to offer insights into the ideal dosage regimen.[Methods] Using specified search phrases, such as “creatine and neurological disorder,” “creatine supplementation and neurodegenerative disorders,” and “creatine and brain,” we searched articles in the PubMed database and Google Scholar. We investigated the association between creatine supplementation and neurodegenerative illnesses by examining references.[Results] The neuroprotective effects of Cr were observed in in vitro and animal models of certain neurodegenerative diseases, while clinical trials failed to reproduce favorable outcomes.[Conclusion] Determining the optimal creatinine regime for increasing brain creatinine levels is essential for maintaining brain health and treating neurodegeneration.
... Research by Chen et al. suggested that L-aspartic acid may affect meat quality by influencing the metabolism of the intestinal microbiota [35]. Additionally, it has been reported that the gene GAMT may improve meat quality by catalyzing creatine synthesis [36,37]. In conclusion, differences in amino acid and energy metabolism likely underlie the higher shear force of Xinglong buffalo meat. ...
Article
Full-text available
Buffalo meat is gaining popularity for its nutritional properties, such as its low fat and cholesterol content. However, it is often unsatisfactory to consumers due to its dark color and low tenderness. There is currently limited research on the regulatory mechanisms of buffalo meat quality. Xinglong buffalo are raised in the tropical Hainan region and are undergoing genetic improvement from draught to meat production. For the first time, we evaluated the meat quality traits of Xinglong buffalo using the longissimus dorsi muscle and compared them to Hainan cattle. Furthermore, we utilized a multi-omics approach combining transcriptomics and metabolomics to explore the underlying molecular mechanism regulating meat quality traits. We found that the Xinglong buffalo had significantly higher meat color redness but lower amino acid content and higher shear force compared to Hainan cattle. Differentially expressed genes (DEGs) and differentially accumulated metabolites (DAMs) were identified, with them being significantly enriched in nicotinic acid and nicotinamide metabolic and glycine, serine, and threonine metabolic pathways. The correlation analysis revealed that those genes and metabolites (such as: GAMT, GCSH, PNP, L-aspartic acid, NADP+, and glutathione) are significantly associated with meat color, tenderness, and amino acid content, indicating their potential as candidate genes and biological indicators associated with meat quality. This study contributes to the breed genetic improvement and enhancement of buffalo meat quality.
... The creatine biosynthetic pathway is crucial for phosphate-related cellular energy production and storage, especially in tissues with high metabolic demands [44]. GAMT is a key enzyme involving the endogenous pathway of creatine biosynthesis and is more expressed in the liver and kidney [45]. Furthermore, glutathione transferase A1 (GSTA1) is a phase II conjugating enzyme that detoxi es electrophilic compounds such as carcinogens, therapeutics, environmental toxins, and oxidative stress products [46]. ...
Preprint
Full-text available
Background: Lupus nephritis (LN) is a severe complication of systemic lupus erythematosus (SLE) with poor treatment outcomes. The role and underlying mechanisms of ferroptosis in LN remain largely unknown. We aimed to explore ferroptosis-related molecular subtypes and assess their prognostic value in LN patients. Methods: Molecular subtypes were classified on the basis of differentially expressed ferroptosis-related genes (FRGs) via the Consensus ClusterPlus package. The enriched functions and pathways, immune infiltrating levels, immune scores, and immune checkpoints were compared between the subgroups. A scoring algorithm based on the subtype-specific feature genes identified by artificial neural network machine learning, referred to as the NeuraLN, was established, and its immunological features, clinical value, and predictive value were evaluated in patients with LN. Finally, immunohistochemical analysis was performed to validate the expression and role of feature genes in glomerular tissues from LN patients and controls. Results: A total of 10 differentially expressed FRGs were identified, most of which showed significant correlation. Based on the 10 FRGs, LN patients were classified into two ferroptosis subtypes, which exhibited significant differences in immune cell abundances, immune scores, and immune checkpoint expression. A NeuraLN-related protective model was established based on nine subtype-specific genes, and it exhibited a robustly predictive value in LN. The nomogram and calibration curves demonstrated the clinical benefits of the protective model. The high-NeuraLN group was closely associated with immune activation. Clinical specimens demonstrated the alterations of ALB, BHMT, GAMT, GSTA1, and HAO2 were in accordance with bioinformatics analysis results, GSTA1 and BHMT were negatively correlated with the severity of LN. Conclusion: The classification of ferroptosis subtypes and establishment of protective model may a foundation for the personalized treatment of LN patients.
Article
Full-text available
Cardiovascular disease remains one of the most common and influential causes of death worldwide, as the heart plays a key role in the human body. Energetic cardioprotective therapy of the myocardium is an important protective factor, regulating the synthesis and use of energy by myocardial cells, while having a little effect on heart rate, blood pressure, and blood supply. Phosphocreatine plays an important role in the intracellular energy buffering and transport system, ensuring energy transfer from the site of production to the site of use to meet the high and variable demands of the heart. Research shows that phosphocreatine can have a positive effect on patients with coronary heart disease, heart failure, and those who have undergone cardiac surgery. The accumulated information confirms the prospects of using phosphocreatine in clinical practice to improve treatment outcomes in patients with heart diseases.
Article
Full-text available
Trichlorfon, one of the most widely used organophosphate insecticides, is commonly employed in aquaculture and agriculture to combat parasitic infestations. However, its inherent instability leads to rapid decomposition into dichlorvos (DDVP), increasing its toxicity by eightfold. Therefore, the environmental effects of trichlorfon in real-world scenarios involve the combined effects of trichlorfon and its degradation product, DDVP. In this study, we systematically investigated the degradation of trichlorfon in tap water over time using HPLC and LC–MS/MS analysis. Subsequently, an experiment was conducted to assess the acute toxicity of trichlorfon and DDVP on goldfish (Carassius auratus), employing a ¹H NMR-based metabolic approach in conjunction with serum biochemistry, histopathological inspection, and correlation network analysis. Exposure to trichlorfon and its degradation product DDVP leads to increased lipid peroxidation, reduced antioxidant activity, and severe hepatotoxicity and nephrotoxicity in goldfish. Based on the observed pathological changes and metabolite alterations, short-term exposure to trichlorfon significantly affected the liver and kidney functions of goldfish, while exerting minimal influence on the brain, potentially due to the presence of the blood–brain barrier. The changes in the metabolic profile indicated that trichlorfon and DDVP influenced several pathways, including oxidative stress, protein synthesis, energy metabolism, and nucleic acid metabolism. This study demonstrated the applicability and potential of ¹H NMR-based metabonomics in pesticide environmental risk assessment, providing a feasible method for the comprehensive study of pesticide toxicity in water environments. Graphical abstract
Article
Full-text available
Methionine is an indispensable amino acid with an important role as the main methyl donor in cellular metabolism for both fish and mammals. Metabolization of methionine to the methyl donor S-adenosylmethionine (SAM) has consequence for polyamine, carnitine, phospholipid, and creatine synthesis as well as epigenetic modifications such as DNA- and histone tail methylation. Methionine can also be converted to cysteine and contributes as a precursor for taurine and glutathione synthesis. Moreover, methionine is the start codon for every protein being synthetized and thereby serves an important role in initiating translation. Modern salmon feed is dominated by plant ingredients containing less taurine, carnitine, and creatine than animal-based ingredients. This shift results in competition for SAM due to an increasing need to endogenously synthesize associated metabolites. The availability of methionine has profound implications for various metabolic pathways including allosteric regulation. This necessitates a higher nutritional need to meet the requirement as a methyl donor, surpassing the quantities for protein synthesis and growth. This comprehensive review provides an overview of the key metabolic pathways in which methionine plays a central role as methyl donor and unfolds the implications for methylation capacity, metabolism, and overall health particularly emphasizing the development of fatty liver, oxidation, and inflammation when methionine abundance is insufficient focusing on nutrition for Atlantic salmon (Salmo salar).
Article
Full-text available
Creatine supplementation is known to exert an effect by increasing strength in high intensity and short duration exercises. There is a hypothesis which suggests that creatine supplementation may provide antioxidant activity by scavenging Reactive Oxygen Species. However, the antioxidant effect of creatine supplementation associated with resistance training has not yet been described in the literature. Therefore, we investigated the effect of creatine monohydrate supplementation associated with resistance training over maximum strength gain and oxidative stress in rats. Forty male Wistar rats (250-300 g, 90 days old) were randomly allocated into 4 groups: Sedentary (SED, n = 10), Sedentary + Creatine (SED-Cr, n = 10), Resistance Training (RT, n = 10) and Resistance Training + Creatine (RT-Cr, n = 10). Trained animals were submitted to the RT protocol (4 series of 10-12 repetitions, 90 second interval, 4 times per week, 65 % to 75 % of 1MR, for 8 weeks). In this study, greater strength gain was observed in the SED-Cr, RT and RT-Cr groups compared to the SED group (P < 0.001). The RT-Cr group showed a higher maximum strength gain when compared to other groups (P < 0.001). Creatine supplementation associated with resistance training was able to reduce lipoperoxidation in the plasma (P < 0.05), the heart (P < 0.05), the liver (P < 0.05) and the gastrocnemius (P < 0.05) when compared to control groups. However, the supplementation had no influence on catalase activity (CAT) in the analyzed organs. Only in the heart was the CAT activity higher in the RT-Cr group (P < 0.05). The activity of superoxide dismutase (SOD) was lower in all of the analyzed organs in the SED-Cr group (P < 0.05), while SOD activity was lower in the trained group and sedentary supplemented group (P < 0.05). Creatine was shown to be an effective non-enzymatic antioxidant with supplementation alone and also when it was associated with resistance training in rats.
Article
Full-text available
Apoptosis is a prominent feature of liver diseases. Causative factors such as alcohol, viruses, toxic bile acids, fatty acids, drugs, and immune response, can induce apoptotic cell death via membrane receptors and intracellular stress. Apoptotic signaling network, including membrane death receptor-mediated cascade, reactive oxygen species (ROS) generation, endoplasmic reticulum (ER) stress, lysosomal permeabilization, and mitochondrial dysfunction, is intermixed each other, but one mechanism may dominate at a particular stage. Mechanisms of hepatic apoptosis are complicated by multiple signaling pathways. The progression of liver disease is affected by the balance between apoptotic and antiapoptotic capabilities. Therapeutic options of liver injury are impacted by the clear understanding toward mechanisms of hepatic apoptosis.
Article
With regard to athletes attempting to improve their performance, at the present time creatine monohydrate is clearly the most widely used dietary supplement or ergogenic aid. Loading doses as high as 20 g/d are typical among athletes. The majority (> 90%) of the creatine ingested is removed from the plasma by the kidney and excreted in the urine. Despite relatively few isolated reports of renal dysfunction in persons taking creatine, the studies completed to date suggest that in normal healthy individuals the kidneys are able to excrete creatine, and its end product creatinine, in a manner that does not adversely alter renal function. This situation would be predicted to be different in persons with impaired glomerular filtration or inherent renal disease. The question of whether long-term creatine supplementation (ie, months to years) has any deleterious affects on renal structure or function can not be answered at this time. The limited number of studies that have addressed the issue of the chronic use of creatine have not seen remarkable changes in renal function. However, physicians should be aware that the safety of long-term creatine supplementation, in regard to the effects on the kidneys, cannot be guaranteed. More information is needed on possible changes in blood pressure, protein/albumin excretion, and glomerular filtration in athletes who are habitual users of this compound.
Article
Anecdotal reports have suggested that creatine supplementation may elevate blood pressure and plasma creatine kinase [CK]. We examined the effects of creatine monohydrate (CR; 20g/d x 5d) and placebo (PL) upon blood pressure (Finapres), plasma [CK], and lean body mass (DEXA). Healthy young females and males (n=30) completed the study in a randomized, double-blind fashion. In addition, an ischaemic handgrip MVC protocol was performed and blood samples were collected at rest and following exercise. There were no treatment effects upon systolic, diastolic, or mean blood pressure. Both total body mass (TBM) and lean body mass (LBM) were increased for the CR vs. PL group (p< .05) with no changes in body fat. Mass changes were greater in males vs. females. Neither resting plasma [CK] nor post-exercise plasma [lactate] were affected by treatment. We concluded that creatine supplementation did not elevate resting blood pressure nor plasma [CK], but increased LBM. Additionally, creatine induced increases in LBM appeared to be greater for males as compared to females.
Article
OBJECTIVE: To examine the effect of creatine supplementation on renal function and estimates of creatinine clearance. DATA SOURCES: A MEDLINE search was conducted (1966-September 2004) using the key terms creatine, creatinine, kidney function tests, drug toxicity, and exercise. Relevant articles were cross-referenced to screen for additional information. DATA SYNTHESIS: Supplementation with creatine, an unregulated dietary substance, is increasingly common in young athletes. To date, few studies have evaluated the impact of creatine on renal function and estimates of creatinine clearance, Because creatine is converted to creatinine in the body, supplementation with large doses of creatine may falsely elevate creatinine concentrations. Five studies have reported measures of renal function after acute creatine ingestion and 4 after chronic ingestion. All of these studies were completed in young healthy populations. Following acute ingestion (4-5 days) of large amounts of creatine, creatinine concentrations increased slightly, but not to a clinically significant concentration. Creatinine is also only minimally affected by longer creatine supplementation (up to 5.6 y). CONCLUSIONS: Creatine supplementation minimally impacts creatinine concentrations and renal function in young healthy adults. Although creatinine concentrations may increase after long periods of creatine supplementation, the increase is extremely limited and unlikely to affect estimates of creatinine clearance and subsequent dosage adjustments. Further studies are required in the elderly and patients with renal insufficiency.
Article
Non-alcoholic fatty liver disease (NAFLD) is the most common liver disease in adults and its prevalence is rising around the world. This pathology is characterized by accumulation of liver fat, which exceeds 5% of liver weight in absence of alcohol consumption, viral infection or other hepatic etiology. Since NAFLD has been associated with obesity, insulin resistance, diabetes or alteration of lipid profiles, it is considered as the liver manifestation of metabolic syndrome. Pathogenic mechanisms of NAFLD have not been clearly elucidated, but different events such as lipid accumulation, insulin resistance, oxidative and endoplasmic reticulum stress, mitochondrial dysfunction and inflammation are involved. Modifications in lifestyle constitute the first line for the management of NAFLD. Nutritional interventions include low fat and carbohydrate diet with higher polyunsaturated fatty acids ingestion. Moreover, supplementation with antioxidant and cytoprotective agents could be useful to decrease oxidative stress, inflammation and fibrosis. Physical activity enables to reduce the expression of lipogenic genes, fat accumulation, or insulin resistance and improves cardiorespiratory fitness. Benefits have been found following both aerobic exercise and resistance training, and remain even after exercise cessation. However, more studies are required to analyze the molecular and cellular mechanisms involved in nutritional and physical intervention, and to define the volume of activity required and its association with weight loss. In this paper, we offer an updated overview of the mechanisms implicated in the progression of NAFLD, and analyze the beneficial effects of nutritional interventions and physical exercise in the prevention and treatment of this condition.
Article
Non-alcoholic fatty liver disease encompasses a wide spectrum of liver damage including steatosis, non-alcoholic steatohepatitis, fibrosis and cirrhosis. We have previously reported that creatine supplementation prevents hepatic steatosis and lipid peroxidation in rats fed a high-fat diet. In this study, we employed oleate-treated McArdle RH-7777 rat hepatoma cells to investigate the role of creatine in regulating hepatic lipid metabolism. Creatine, but not structural analogues, reduced cellular TG accumulation in a dose-dependent manner. Incubating cells with the pan-lipase inhibitor diethyl p-nitrophenylphosphate (E600) did not diminish the effect of creatine, demonstrating that the TG reduction brought about by creatine does not depend on lipolysis. Radiolabeled tracer experiments indicate that creatine increases fatty acid oxidation and TG secretion. In line with increased fatty acid oxidation, mRNA analysis revealed that creatine-treated cells had increased expression of PPARα and several of its transcriptional targets. Taken together, this study provides direct evidence that creatine reduces lipid accumulation in hepatocytes by the stimulation of fatty acid oxidation and TG secretion.
Article
Creatine has become a popular nutritional supplement among athletes. Recent research has also suggested that there may be a number of potential therapeutic uses of creatine. This paper reviews the available research that has examined the potential ergogenic value of creatine supplementation on exercise performance and training adaptations. Review of the literature indicates that over 500 research studies have evaluated the effects of creatine supplementation on muscle physiology and/or exercise capacity in healthy, trained, and various diseased populations. Short-term creatine supplementation (e.g. 20 g/day for 5–7 days) has typically been reported to increase total creatine content by 10–30% and phosphocreatine stores by 10–40%. Of the approximately 300 studies that have evaluated the potential ergogenic value of creatine supplementation, about 70% of these studies report statistically significant results while remaining studies generally report non-significant gains in performance. No study reports a statistically significant ergolytic effect. For example, short-term creatine supplementation has been reported to improve maximal power/strength (5–15%), work performed during sets of maximal effort muscle contractions (5–15%), single-effort sprint performance (1–5%), and work performed during repetitive sprint performance (5–15%). Moreover, creatine supplementation during training has been reported to promote significantly greater gains in strength, fat free mass, and performance primarily of high intensity exercise tasks. Although not all studies report significant results, the preponderance of scientific evidence indicates that creatine supplementation appears to be a generally effective nutritional ergogenic aid for a variety of exercise tasks in a number of athletic and clinical populations.