You are on page 1of 5

Brain Activation in Uremic Anorexia

Abelardo Aguilera, MD, José Antonio Sánchez-Tomero, MD, PhD,


and Rafael Selgas, MD, PhD

This article reviews current knowledge about mechanisms responsible for uremic events, especially those that
involve the central nervous system (CNS). Anorexia is a frequent complication of the uremic syndrome that
contributes to malnutrition in patients on dialysis. Uremic anorexia has been associated with many factors.
Traditionally, anorexia in dialysis patients has been regarded as a sign of uremic toxicity; therefore, 2 hypotheses
have been proposed: the “middle molecule” and “peak concentration” hypotheses; both of these remain unproved.
Recently, our group has proposed the tryptophan-serotonin hypothesis, which is based on a disorder in the amino
acid profile that may be acquired when the patient is in uremic status. It is characterized by low concentrations of
large neutral and branched chain amino acids in the cerebrospinal fluid. This situation permits a high level of
tryptophan transport across the blood– brain barrier and enhances the synthesis of serotonin (the final target
responsible for inhibiting appetite). The role of inflammation in the genesis of anorexia–malnutrition is also
emphasized. In summary, in the CNS, factors associated with uremic anorexia include high levels within the
cerebrospinal fluid of proinflammatory cytokines, leptin, and free tryptophan and serotonin (hyperserotoninergic-
like syndrome), along with deficiency of neural nitric oxide (nNO) and disorders in various receptors such as
melanocortin receptor-4 (MC4-R). Uremic anorexia is a complex complication associated with malnutrition and high
levels of morbidity and mortality. Several uremia-acquired disorders in the CNS such as high cerebrospinal fluid
levels of anorexigen substances and disorders in appetite regulator receptors may explain the lack of appetite.
© 2007 by the National Kidney Foundation, Inc.

A NOREXIA IS A FREQUENT symptom of


uremic syndrome that contributes to mal-
nutrition in patients on dialysis. The clinical rel-
knowledge about the mechanisms responsible for
uremic anorexia, especially disorders involving
the central nervous system (CNS).
evance of anorexia and malnutrition in patients Regulation of food intake involves a heteroge-
on dialysis is related to associated levels of mor- neous feedback system that requires the participa-
bidity, mortality, and hospitalization.1,2 Tradi- tion of various signals derived from gastrointestinal
tionally, anorexia has been considered a sign of tract (GIT), liver, circulating nutrients, fat stores,
uremic intoxication; it has been used as a clinical and metabolizing cell products, all of which transmit
marker to initiate dialysis or define it as inade- information to the CNS. Inputs from the peripheral
quate.1 In dialysis patients, appetite regulation is nervous system, including the vagus nerve and its
complicated for the abnormal metabolism and gastric receptors, are also important. Signals are
renal retention of several substances; effects on integrated into the CNS, thereby inducing hunger
the hunger–satiety cycle have been observed. or satiety. Within the CNS, the hypothalamus con-
Frequently, in uremic patients, this equilibrium is tains the feeding center (lateral hypothalamus) with
influenced to induce anorexia through the pre- dopaminergic receptors, along with the satiety cen-
dominant renal retention of peptides and cyto- ter (ventromedial hypothalamus) with serotoniner-
kines.3 However, the exact cause of uremic an- gic and adrenergic neurotransmitters.4 Two impor-
orexia is unknown. This paper reviews current
tant pathways are included in the hypothalamus:
melanocortin receptor-4 (MC4-R) and pro-
Servicio de Nefrologia, Hospitales Universitarios de la Princesa y opimelanocortin (POMC) stimulation.5 Under
la Paz, Madrid, Spain. normal conditions, the activity of MC4-R is in-
Address reprint requests to Abelardo Aguilera, MD, Servicio de creased in 2 ways: through suppression of orexigen
Nefrologia, Hospitales Universitarios de la Princesa, Diego de Leon hormones such as neuropeptide Y (NPY) and ag-
62, 28046 Madrid, Spain. E-mail: aguileraa@terra.es
© 2007 by the National Kidney Foundation, Inc.
outi-relative protein–like (AGRP) activation of
1051-2276/07/1701-0010$32.00/0 POMC-expressing neurons to produce ␣-melano-
doi:10.1053/j.jrn.2006.10.020 cyte–stimulating hormone (␣-MSH). This directly

Journal of Renal Nutrition, Vol 17, No 1 ( January), 2007: pp 57-61 57

Descargado para Anonymous User (n/a) en Universidad Nacional Autonoma de Mexico de ClinicalKey.es por Elsevier en julio 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
58 AGUILERA ET AL

BLOOD HYPOTHALAMUS
UREMIA
CCK Ghrelin Leptin L-arginine NO
1
Renal acidosis NPY
Liquid overload nNOS / ADMA

TNF- 2 Serotonin
CSF TRP Dopamine /
Plasma TRP Serotonin ratio
(BCAA+PHE+TYP+MET)
TRH Catecholamines
Catecholamines 4
Insulin AgRP -MSH

Leptin
5 MC4-Rs
L-arginine NO NPY
3

Blood
NOS / ADMA
TNF- Brain ANOREXIA
Barrier
Figure 1. Factors implicated in the pathogenesis of uremic anorexia and its treatment. CCK, cholecystokinin;
TNF-␣, tumor necrosis factor-alpha; TRP, tryptophan; BCAA, branched chain amino acid; NO, nitric oxide; NPY,
neuropeptide Y; NOS, nitric oxide synthase; nNOS, neural nitric oxide synthase; ADMA, asymmetric dimethyl-
L-arginine; CSF, cerebrospinal fluid; TRH, tryptophan hydroxylase; AgRP, agouti-related protein; ␣-MSH,
alpha-melanocyte stimulating hormones; PHE, phenylalanine; TYP, tyrosine phosphatase; MET, methionine.
In the treatment of patients with uremic anorexia, the numbers indicate the specific sites of action: 1, CCK
antagonist; 2, anti-TNF-␣ drugs (specific antibodies, megestrol acetate, thalidomide, others); 3, increased
NPY (megestrol acetate); 4, inhibitors of tryptophan transport across the blood– brain barrier (large neutral
amino and branched chain acid inhibitors); and 5, blocking of melanocortin receptor-4 (MC4-R).

stimulates MC4-R, causing increased energy ex- and central appetite control. During gastric and
penditure and reduced food intake.5 High plasma preabsorptive phases, changes in the plasma
levels of insulin, leptin, peptide YY3-36 (PYY3-36) amino acid profile influence peripheral food in-
from the colon, and possibly other uremic toxins take. Additionally, contact between dietary
induce MC4-R stimulation (Fig 1).6 amino acids and GIT receptors stimulates CCK
Signals from GIT and liver reach the hypothal- release.13 In the postabsorptive phase, changes in
amus through 2 routes: through the peripheral plasma amino acid concentrations also induce
nerves (mainly the vagus) and via the bloodstream satiety.7 Centrally, some amino acids can act as
as carbohydrates, amino acids, peptide, hor- neurotransmitters or precursors of a neurotrans-
mones, nitric oxide (NO), and cytokines. Two mitter, inducing hunger or satiety.14 Catechol-
main neuropeptides, cholecystokinin (CCK) and amine and serotonin pathways appear to be in-
NPY, have been implicated in central appetite volved in the hypothalamic regulation of feeding.
control. CCK is a recognized anorexigen agent,7 Catecholamine synthesis is influenced by brain
and NPY is the most potent orexigen known.8 levels of its precursor, tyrosine. Serotonin is syn-
Cytokines (tumor necrosis factor [TNF]-␣ and thesized in the brain from its precursor, trypto-
interleukin [IL]-1) are released during infection phan. Increasing brain serotonin activity is asso-
or malignant processes9 and are accumulated in ciated with reduced food intake. In the regulation
dialysis patients.10 Nine TNF-␣ molecular frag- of eating behavior, 2 factors play a pivotal role—
ments have been isolated, each with a different concentration of free tryptophan (not bound to
action. The 69 –100 fragment has suppressive albumin) (FTRP) and molar ratio between
effects on food intake.11 IL-1 may also contribute FTRP and other plasma large neutral amino acids
to anorexigen effects of CCK and TNF-␣.12 (LNAAs). LNAAs and branched chain amino
On the other hand, substances such as brain acids (BCCAs) are able to compete with trypto-
amino acids and NO are implicated in peripheral phan for brain entry.14

Descargado para Anonymous User (n/a) en Universidad Nacional Autonoma de Mexico de ClinicalKey.es por Elsevier en julio 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
BRAIN ACTIVATION IN UREMIC ANOREXIA 59

Table 1. Possible Disorders in Brain Appetite Cytokines and Uremic Anorexia


Regulation in Uremic Patients
IL-1 and TNF-␣ have been shown to interfere
● Disturbances in the peripheral and autonomic nervous
with tryptophan and serotonin turnover. It is
system (axonal vagus nerve degeneration). The vagus
nerve is responsible for transporting satiety or feeding therefore possible that, by interfering with the
sensation from the gastrointestinal tract to the brain. mechanism that regulates the plasma tryptophan
● High plasma levels of anorexigen substances able to pool or by increasing brain serotonin release, IL-1
cross the blood– brain barrier and induce anorexia per and TNF-␣ may participate in uremic serotonin-
se. Peptides include CCK, glucagon, insulin, leptin,
induced anorexia.9,14 In support of this hypoth-
cortisol release factor, pancreatic polypeptide,
␣-MSH, and PYY3-36. Cytokines (TNF-␣ and IL-1) esis, other groups10 have reported high TNF-␣
lower zinc and possibly NPY.* plasma levels in patients on peritoneal dialysis
● Lowered nNO synthesis through decreased nNOS (PD). TNF-␣ plasma levels were significantly
associated with accumulation of ADMA and high higher in patients with anorexia and malnutri-
levels of TNF-␣.
tion.18 Moreover, TNF-␣ is able to inhibit he-
● Inactivation of TPH by NO deficiency and increased
FTRP. patic albumin gene expression, thereby contrib-
● High levels of FTRP (producing increased serotonin uting to dialysis protein malnutrition.19
synthesis).
● Imbalance in brain amino acids (high
FTRP/LNAA-BCCA ratio). NO and Uremic Anorexia
● Sympathetic hyperactivity with high levels of Accumulation of endogenous NO inhibitors
dopamine, norepinephrine, and serotonin.
● Disturbances in the regulation of MC4-R. (asymmetric dimethyl-L-arginine [ADMA]) has
been described in uremic patients.20 Experimen-
Abbreviations: CCK, cholecystokinin; ␣-MSH, alpha-mel-
anocyte stimulating hormone; PYY3-36, peptide YY3-36;
tally, the administration of L-NAME increases
TNF-␣, tumoral necrosis factor-alpha; IL-1, interleukin-1; blood pressure and brain serotonin concentration,
NPY, neuropeptide Y; nNO, neural nitric oxide; nNOS, neu- induces gastric distention, and promotes and fa-
ral NO synthase; ADMA, asymmetric dimethyl-L-arginine; cilitates oxidation of L-arginine to ornithine.15
TPH, tryptophan hydroxylase; FTRP, free tryptophan; LNAA, Also, TNF-␣ decreases NOS levels through se-
plasma large neutral amino acid; BCCA, branched chain
amino acid; MC4-R, melanocortin receptor-4.
lective destabilization of mRNA transcription,
*Other groups found high plasma levels. In our study,27 and it enhances sympathetic activity, thus induc-
NPY was normal in 80% of cases, and relatively low ing anorexia again.21 Therefore, uremic NO de-
values were found in anorexic patients on peritoneal ficiency via inactivation of brain TPH may be
dialysis. another cause of anorexia.16

Leptin and Uremic Anorexia


NO, an endothelium-released factor synthe-
Leptin, a 16-kDa protein that is produced by
sized from the amino acid L-arginine by a specific
adipocytes, regulates body fat through a central
synthase (nitric oxide synthase [NOS]), partici-
satiety effect. Hyperleptinemia, possibly caused
pates in brain development and function. Exper-
by uremic retention, hyperproduction, or both,
imentally, the administration of NG-nitro-L-ar- has been described in cases of uremia. It has been
ginine methyl ester (L-NAME), an inhibitor of suggested that leptin may be involved in malnu-
NOS, reduces food intake in rats.15 Interesting trition of dialysis patients. A leptin receptor dis-
observations have shown an interaction between order has been noted in an obese population, and
brain NO levels and serotonin synthesis.15 Brain acquired leptin receptor disorders (ie, resistance
NO inactivates brain tryptophan hydroxylase to insulin or parathormone) may be speculated in
(TPH), decreasing brain tryptophan and subse- uremic individuals. Uremic patients may also
quently serotonin and catecholamine levels. Both have normal sensibility to the hypothalamic lep-
dopamine and norepinephrine increase the avail- tin receptor. As a consequence, the uremic hy-
ability of brain tryptophan, initiating a vicious perleptinemia may up-regulate the hypothalamic
circle.16 In uremia, NO deficiency has also been leptin receptor. The mechanism by which leptin
implicated in the pathogenesis of cardiovascular induces appetite suppression is reducing the hy-
complications.17 Table 1 lists the CNS disorders pothalamic NPY levels and increasing the sym-
that are responsible for uremic anorexia. pathetic activity with hyperinsulinaemia.22 Con-

Descargado para Anonymous User (n/a) en Universidad Nacional Autonoma de Mexico de ClinicalKey.es por Elsevier en julio 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
60 AGUILERA ET AL

sistent with this hypothesis is the sympathetic proinflammatory molecules (IL-1 and TNF-␣)
hyperactivity (ie, dopamine, norepinephrine, se- accumulated through renal failure or overpro-
rotonin) described in uremic patients.23 More- duced by inflamed tissues or overhydration28
over, in rats, injection of leptin decreases brain augment renal acidosis, hyperinsulinemia, sympa-
nNOS, thyrotropin-releasing hormone, and cor- thetic activity, and FTRP, with decreases re-
ticotropin-releasing hormone mRNA in para- ported in LNAA and NOS activity. High CCK
ventricular and supraoptic nuclei, with conse- plasma levels reveal a synergistic anorexigen effect
quent increases in diencephalic serotonin with IL-1, caused by direct induction of decreases
content.24 Leptin also acts as a proinflammatory in LNAA (Fig 1).
molecule.22 Recently, a very important disorder at the end
of the hunger–satiety pathway (MC4-R) has
been described in uremic mice with leptin recep-
Plasma and Brain Amino Acids tor deficiency (db/db) and in MC4-R knockout
and Uremic Anorexia mice (MC4-RKO).29 Cheung and colleagues29
Substantial evidence suggests that changes in injected AGRP into mice with subtotal nephrec-
plasma amino acid profile affect brain or neuro- tomy in the lateral ventricle, inducing increased
transmitter synthesis.14 A significant change in food intake and body growth and a decline in
unbound plasma tryptophan and/or an increase in resting metabolic rate, which suggests that uremia
tryptophan competing amino acids results in se- at particular stages may cause a defect in the
rotonergic hyperactivity and anorexia. These ob- ability of AGRP to block MC4-Rs. Another
servations have contributed to the tryptophan– important conclusion of this article is that leptin,
serotonin hypothesis, which has been proved in by signaling its receptor, is an important cause of
rats.14 Changes in the central amino acid profile, uremic cachexia; uremic mice with leptin recep-
which have been observed in uremic humans14,25 tor deficiency (db/db) grew in a similar manner to
and rats, consist of low glutamine and ␥-ami- sham-operated, pair-fed (db/db) mice. However,
nobutyric acid (GABA); high levels of glycine it is impossible to exclude the possibility that
and free tryptophan (with reduced total plasma other mechanisms such as acidosis and inflamma-
tryptophan); elevated phenylalanine/tyrosine ra- tion may act in an independent manner, thereby
tio; increases in 5-hydroxyindoleacetic and ho- inducing cachexia. All these findings should be
movanillic acids and their ratio; and increased connected to the tryptophan–serotonin disorder
levels of alanine, lysine, arginine, histidine, hypothesis proposed by our group.30 We believe
3-methyl-histidine, citrulline, and ornithine.25,26 that evidence implies a connection between the
Evidence favors the possibility that hypothalamic brain hyperserotoninergic-like syndrome or tryp-
serotonin has a greater effect in reducing carbo- tophan–serotonin disorder hypothesis and the
hydrates with respect to protein or fat ingestion. disorders described by Cheung et al29:
Moreover, long-term serotonin administration
induces an important reduction in all macronu- 1. Serotonin is the most important regulator
trients under experimental conditions.27 of MC4-R expression. In fact, serotonin
High plasma and intracerebral concentrations reuptake inhibitors and MC4-R antago-
of FTPR and resultant increases in serotonin nists share antidepression and antianxiety
enhance the serotonin/dopamine ratio. This phe- effects.31
nomenon may be perpetuated through high con- 2. Serotonin shows an acute and direct effect
centrations of IL-1, TNF-␣, CCK, leptin, cat- on AGRP release, favoring MC4-R over-
echolamines, and insulin; by metabolic acidosis; expression and causing anorexia.32
and by low levels of NO induced by high ADMA 3. Ghrelin, an orexigen stomach hormone,
concentration (Fig 1). Leptin induces hyperinsu- provides negative feedback to brain
linemia by causing insulin resistance, sympathetic serotonin.33
hyperactivity, and decreased NPY, whereas NPY In summary, all these systems are confluent in
decreases NOS activity and NO production. Low inducing and/or perpetuating hyperserotoniner-
NO concentrations induce an increase in TPH, gic brain status, which translates into anorexia.
and consequently, an increase in FTRP, with Uremic anorexia is a complex complication
decreases in LNAA noted. On the other hand, associated with malnutrition and high levels of

Descargado para Anonymous User (n/a) en Universidad Nacional Autonoma de Mexico de ClinicalKey.es por Elsevier en julio 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.
BRAIN ACTIVATION IN UREMIC ANOREXIA 61

morbidity and mortality. Several uremia-acquired 17. Ritz E, Villance P, Nowicki M: The effect of malnutri-
disorders in the CNS such as high cerebrospinal tion on cardiovascular mortality in dialysis patients: Is L-arginine
the Answer? Nephol Dial Transplant 9:129-130, 1994
fluid levels of anorexigen substances and disorders 18. Aguilera A, Codoceo R, Selgas R, et al: Anorexigen
in appetite regulator receptors may explain the (TNF-␣, cholecystokinin) and orexigen (neuropeptide Y) plasma
lack of appetite. These recent findings open pos- levels in peritoneal dialysis (PD) patients: Their relationship with
sibilities for new therapeutic targets. nutritional parameters. Nephrol Dial Transpl 13:1476-1483,
1998
19. Brenner D, Buck M, Feitelberg S, et al: Tumor necrosis
References factor alpha inhibits albumin gene expression in a murine model
1. Bergström J: Appetite in CAPD patients. Perit Dial 16: of cachexia. J Clin Invest 85:248-255, 1990
S181-S184, 1996 (suppl 1) 20. Vallance P, Leone A, Calver A, et al: Accumulation of an
2. Owen W, Lew N, Luí Y, et al: The urea reduction ratio endogenous inhibitor nitric oxide synthesis in chronic renal
and serum albumin concentration as predictors of mortality in failure. Lancet 339:572-575, 1992
patients undergoing hemodialysis. N Engl J Med 329:1001-1006, 21. Ando T, Dunn AJ: Mouse tumor necrosis factor alpha
1993 increases brain tryptophan concentrations and norepinephrine
3. Aguilera A, Selgas R, Diéz JJ, et al: Anorexia in end-stage metabolism while activating the HPA axis in nice. Neuroimmu-
renal disease: Pathophysiology and treatment. Expert Opin Phar- nomodulation 6:319-329, 1999
macother 2:1825-1838, 2001 22. Stenvinkel P: Leptin—A new hormone of definite interest
4. Stricker EM: Biological bases of hunger and satiety: Ther- for the nephrologist. Nephrol Dial Transplant 13:1099-1101, 1998
apeutic implications. Nutr Rev 42:333-340, 1984 23. Converse RL, Jacobsen TN, Toto RD, et al: Sympha-
5. Butler AA, Marks DL, Fan W, et al: Melanocortin-4 thetic overactivity in patients with chronic renal failure. N Engl
receptor is required for acute homeostatic responses to increase J Med 327:1912-1918, 1992
dietary fat. Nat Neurosci 4:605-611, 2001 24. Isse T, Ueta Y, Serino R, et al: Effects of leptin on
6. Batterham RL, Cowley MA, Small CJ, et al: Gut hormone fasting-induced inhibition of neural nitric oxide synthase mRNA
PYY3-36 physiologically inhibits food intake. Nature 418:650- in paraventricular and supraoptic nuclei of rats. Brain Res 846:
654, 2002 229-235, 1999
7. Anderson GH, Li ETS, Anthony SP, et al: Dissociation 25. Laviano A, Cangiano C, Preziosa I, et al: Plasma trypto-
between plasma and brain amino acid profiles and short-term phan and anorexia in liver cirrhosis. Int J Eating Disord 21:181-
food intake in the rat. Am J Physiol 35:R1675-R1686, 1994 186, 1996
8. Dube MG, Xu B, Crowley WR, et al: Evidence that 26. Siassi F, Wang M, Kopple JD, et al: Plasma tryptophan
neuropetide Y is a physiological signal of normal food intake. levels and brain serotonin metabolism in chronically uremic rats.
Brain Res 646:341-344, 1994 J Nutr 107:840-845, 1977
9. Plata-Salamán C: Cytokines and feeding suppression: An 27. Leibowitz SF, Alexander JT: Hypothalamic serotonin in
integrative view from neurologic to molecular levels. Nutrition control of eating behavior, meal size, and body weight. Biol
11:674-677, 1995 (suppl 5) Psychiatry 44:851-864, 1998
10. MacDonald C, Rush D, Bernstein K, et al: Production of 28. Niebauer J, Volk HD, Kemp M, et al: Endotoxin and
tumoral necrosis factor in hemodialysis. Nephron 65:273-277, immune activation in chronic heart failure: A prospective cohort
1993 study. Lancet 353:1838-1842, 1999
11. Kapás L, Hong L, Cady AB, et al: Somnogenic, pyro- 29. Cheung W, Yu PX, Little BM, et al: Role of leptin and
genic, and anorectic activities of tumor necrosis factor alpha and melanocortin signalling in uremia-associated cachexia. J Clin
TNF-␣ fragments. Am J Physiol 263:R708-R715, 1993 Invest 115:1659-1665, 2005
12. Treget EE, Yu YM, Zhong S, et al: Role of interleukin 1 30. Aguilera A, Codoceo R, Bajo MA, et al: Eating behavior
and tumor necrosis factor in energy metabolism in rabbits. Am J disorders in uremia: A question of balance in appetite regulation.
Physiol 255:E760-E768, 1988 Semin Dial 17:44-52, 2004
13. Bellinger LL, Williams FE, Rogers QR, et al: Liver 31. Chaki S, Hirota S, Funakoshi T, et al: Anxiolytic-like and
denervation attenuate the hypophagia produced by an imbal- antidepressant-like activities of MCLO129 (1-((S)-2-(4-fluoro-
anced acid diet. Physiol Behav 59:925-929, 1996 phenyl)-2-(4-isopropylpiperadin-1-yl) ethyl)-4-(2-methoxynaph-
14. Cangiano C, Laviano A, Muscaritoli M, et al: Cancer thalen-1-yl) butyl) piperazine), a novel and potent nonpeptide
anorexia: New pathogenic and therapeutic insights. Nutrition antagonist of the melanocortin-4 receptor. J Pharmacol Exp Therap
12:S48-S50, 1996 304:818-826, 2003
15. Squadrito F, Calapai D, Altavilla D, et al: Central serotoner- 32. Li JY, Finniss S, Yang YK, et al: Agouti-related protein–
gic system involvement in the anorexia induced by NG-nitro-L- like immunoreactivity: Characterization of release from hypo-
arginine, an inhibitor of nitric oxide synthase. Eur J Pharmacol thalamic tissue and presence in serum. Endocrinology 141:1942-
255:51-55, 1994 1950, 2000
16. Kuhn DM, Arthur R: Molecular mechanism of the inac- 33. Nonogaki K, Ohashi-Nozue K, Oka Y: A negative feed-
tivation of tryptophan hydroxylase by nitric oxide: Attack on back system between brain serotonin systems and plasma active
critical sulfhydryls that spare the enzyme iron center. J Neurosci ghrelin levels has been described in mice. Biochem Biophys Res
17:7245-7251, 1997 Commun 341:703-707, 2006

Descargado para Anonymous User (n/a) en Universidad Nacional Autonoma de Mexico de ClinicalKey.es por Elsevier en julio 04, 2018.
Para uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2018. Elsevier Inc. Todos los derechos reservados.

You might also like