You are on page 1of 7

Send Orders for Reprints to reprints@benthamscience.

net
Current Pharmaceutical Design, 2019, 25, 1-7 1
RESEARCH ARTICLE

Activation of Melanocortin-4 Receptor by a Synthetic Agonist Inhibits Ethanol-


induced Neuroinflammation in Rats

Osvaldo Flores-Bastíasa,b, Gonzalo I. Gómeza,b, Juan A. Orellanab,c and Eduardo Karahaniana,b*

a
Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud,, Universidad Autónoma de Chile, Chile; bResearch Center for
the Study of Alcohol Drinking Behavior in Adolescents, Santiago, Chile; cDepartamento de Neurología, Escuela de Medicina and
Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile

Abstract: Background: High ethanol intake induces a neuroinflammatory response resulting in the subsequent
maintenance of chronic alcohol consumption. The melanocortin system plays a pivotal role in the modulation of
alcohol consumption. Interestingly, it has been shown that the activation of melanocortin-4 receptor (MC4R) in
the brain decreases the neuroinflammatory response in models of brain damage other than alcohol consumption,
such as LPS-induced neuroinflammation, cerebral ischemia, glutamate excitotoxicity, and spinal cord injury.
Objectives: In this work, we aimed to study whether MC4R activation by a synthetic MC4R-agonist peptide
prevents ethanol-induced neuroinflammation, and if alcohol consumption produces changes in MC4R expression
in the hippocampus and hypothalamus.
ARTICLE HISTORY
Methods: Ethanol-preferring Sprague Dawley rats were selected offering access to 20% ethanol on alternate days
for 4 weeks (intermittent access protocol). After this time, animals were i.p. administered an MC4R agonist pep-
Received: October 30, 2019 tide in the last 2 days of the protocol. Then, the expression of the proinflammatory cytokines interleukin 6 (IL-6),
Accepted: December 10, 2019
interleukin 1-beta (IL-1β), and tumor necrosis factor-alpha (TNF-α) were measured in the hippocampus, hypo-
thalamus and prefrontal cortex. It was also evaluated if ethanol intake produces alterations in the expression of
DOI: MC4R in the hippocampus and the hypothalamus.
10.2174/1381612825666191216145153  
Results: Alcohol consumption increased the expression of MC4R in the hippocampus and the hypothalamus. The
administration of the MC4R agonist reduced IL-6, IL-1β and TNF-α levels in hippocampus, hypothalamus and
prefrontal cortex, to those observed in control rats that did not drink alcohol.
Conclusion: High ethanol consumption produces an increase in the expression of MC4R in the hippocampus and
hypothalamus. The administration of a synthetic MC4R-agonist peptide prevents neuroinflammation induced by
alcohol consumption in the hippocampus, hypothalamus, and prefrontal cortex. These results could explain the
effect of α-MSH and other synthetic MC4R agonists in decreasing alcohol intake through the reduction of the
ethanol-induced inflammatory response in the brain.
Keywords: MC4R, melanocortin, α-MSH, alcohol use disorder, neuroinflammation.

1. INTRODUCTION maintenance of chronic alcohol consumption and (iii) alcohol re-


According to the World Health Organization (WHO), alcohol is lapse behavior, the notion that neuroinflammation and oxidative
the most commonly abused drug and its uncontrolled consumption, stress would play a role in the establishment of chronic alcohol
called alcohol use disorder (AUD), is among the top five risk fac- abuse has gained importance recently [11]. An interesting hypothe-
tors for disease and death worldwide [1, 2]. Indeed, a study in the sis is that neuroinflammation plays a critical role in the establish-
United Kingdom concluded that alcohol is the most harmful drug to ment of addiction to several substances of abuse, including alcohol
the society, surpassing heroin and cocaine [3]. Alcohol drinking is [12, 13], particularly, through the activation of genes related to the
associated with a risk of developing health problems such as cancer, innate immune system. Chronic activation of the innate immune
liver cirrhosis and various disabilities, in addition to an increased response in the brain would produce long-lasting neurobiological
risk of accidents [4, 5]. Data from the WHO indicate that the global changes, e.g., increased glutamate-induced hyperexcitability and
effect of noxious use of alcohol is approximately 3.3 million deaths excitotoxicity, decreased neurogenesis and behavioral changes such
each year; thus, it accounts for 5.9% of all deaths worldwide [1]. as anxiety, negative affect, and depression, all of which are related
to addictive substances abuse [14].
Along with the various health problems mentioned above, the
excessive consumption of alcohol generates dramatical conse- Microglia and astrocytes are the major sources of inflammatory
quences in the brain such as neurodegeneration [6] and deteriora- mediators and ROS in the nervous system and highly sensitive to
tion of the prefrontal cortex that leads to impaired executive func- ethanol exposure [15]. Upon ethanol stimulation, glial cells release
tion (planning, thinking, and judgment) [7], depression [8], anxiety relevant amounts of proinflammatory cytokines, ROS and nitric
[9] and memory impairment [10]. Although there are multiple neu- oxide (NO) - all of them crucial for the brain inflammatory re-
robiological mechanisms that account for the various aspects re- sponse [16]. Within the brain, the hippocampus is one of the most
lated to AUD, i.e. (i) the acquisition of alcohol intake, (ii) the pathologically affected regions during chronic ethanol intake [17,
18]. The presence of reactive astrocytes and microglia in the hippo-
*Address correspondence to this author at the Institute of Biomedical Sci-
campus are exacerbated in alcoholics [19]. This glial long-lasting
ences, Faculty of Health Sciences, Universidad Autónoma de Chile, Llano process promotes neuronal death in the hippocampus and other
Subercaseaux 2801, San Miguel, 8910060, Santiago, Chile; brain regions via activation of inflammatory routes that cause exci-
Tel: +56-2-23036664; E-mail: eduardo.karahanian@uautonoma.cl totoxicity and ROS production [20]. Microglia, astrocytes and neu-
rons abundantly express NF-κB, a pivotal transcription factor in the

1381-6128/19 $58.00+.00 © 2019 Bentham Science Publishers


2 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Flores-Bastías et al.

genesis and progression of the brain of innate immune-response is highly expressed in both microglia and astrocytes [44, 45, 46],
genes. NF-κB is involved in ethanol-induced neuroinflammation which suggests that α-MSH may exert its anti-inflammatory effects
[21] and can be directly activated by ROS [22]. Likewise, ethanol via MC4Rs in these key neuroinflammatory cells [47]. In fact, the
increases cytochrome P4502E1 (Cyp2E1) activity [23], and in turn, impact of MC4R activation on the reduction of neuroinflammation
Cyp2E1 metabolism of ethanol increases ROS levels resulting in has been studied in other neuropathological models of brain injury
the NF-κB-mediated production of proinflammatory mediators such different from ethanol intake: experimental brain inflammation
as IL-6, IL-1β and TNF-α [28]. In addition, NF-κB activation pro- induced by LPS [48, 49, 50], cerebral ischemia damage [51, 52],
duces a greater expression of NADPH oxidase [24], an enzyme that kainic acid-induced excitotoxic damage [53] and experimental spi-
produces even more ROS, which creates a positive activation loop nal cord injury [54, 55]. In all these studies, the neuroprotective
that rapidly potentiates innate immune response [13, 14]. In accor- actions of α-MSH and other synthetic analog peptides reside in their
dance with this, the daily administration of N-acetyl cysteine (a ability to decrease NF-κB signaling in glial cells, which is the key
precursor in the formation of the antioxidant glutathione) to rats transcription pathway for the induction of innate immune-response
that have chronically consumed ethanol markedly reduces voluntary [13]. Specifically, α-MSH and other synthetic melanocortin recep-
ethanol intake [11]. In addition to the intrinsic neuronal death medi- tor ligands prevent IκBα phosphorylation and, consequently, inhibit
ated by ROS, activation of NF-κB signaling stimulates the release NF-κB activation [56]. In agreement, α-MSH also decreases the
of TNF-α, IL-1β, IL-6, and transforming growth factor-β (TGF-β1), production of key pro-inflammatory cytokines such as TNFα [51,
resulting in neuronal apoptosis [25]. A persistent status of neuroin- 52, 53, 57, 58, 59], enzymes such as iNOS and COX-2 [47] and
flammation, together with the exacerbated production of ROS, diverse inflammatory mediators, like NO [60, 61] in the brain.
would lead to neurodegeneration of key areas involved in alcohol Along with the inhibition of NF-κB activity, α-MSH also stimulates
consumption and addiction [13]. the release of the anti-inflammatory cytokines IL-10 and TGF-β
It has been proposed that, instead of simply being a side effect from microglia and astrocytes, respectively, by a mechanism in-
of excessive alcohol consumption, neuronal damage associated with volving the expression of the peroxisome proliferator-activated
drinking may actually underlie some of the mechanisms that regu- receptor gamma (PPARγ) [62].
late the development of AUD [25]. Alcohol-induced cell death in Given that neuroinflammation underlies the progression of chronic
regions such as the prefrontal cortex may lead to a lack of inhibition alcohol intake, and because brain MC4R activation has anti-
in subcortical reward areas like the striatum, which in turn may inflammatory effects, we investigated whether heavy alcohol con-
reduce behavioral inhibition and increase motivation to drink. Re- sumption induces neuroinflammation and alteration in MC4R ex-
peated stimulation of the innate immune system during chronic or pression in brain areas susceptible to the greatest effects of ethanol-
heavy alcohol consumption may facilitate this process, leading to induced neuroinflammation. Additionally, we evaluated if the acti-
decreased inhibition of the mesolimbic reward system and thus, vation of MC4R by a selective synthetic agonist-peptide prevents
increasing drinking [14, 29]. the alcohol-induced neuroinflammatory response.
The hypothalamus is one of the brain regions that undergo more
2. MATERIALS AND METHODS
physiological changes during excessive alcohol intake [26]. Within,
several orexigenic peptides (such as orexin, encephalin andgalanin) 2.1. Animals, Ethanol Consumption and Treatment with an
stimulate alcohol consumption [27, 28, 29]; on the other hand, MC4R Agonist
anorexigenic peptides including corticotropin-releasing factor, the
endogenous opioid dynorphin and melanocortins, inhibit alcohol The experiments were performed in male Sprague Dawley out-
drinking [29, 30, 31]. Melanocortins (MCs) belong to a group of bred rats purchased from the Pontificia Universidad Católica de
neuropeptides that are produced in the arcuate nucleus of the hypo- Chile animal facility. Rats were housed in individual cages in a
thalamus (Arc) which include the α-, β-, and γ- melanocyte- temperature-controlled room on a reversed 12-h light/12-h dark
stimulating hormones (α-MSH, β-MSH and γ-MSH) [32]. α-MSH cycle, with food and water provided ad libitum. Twenty-five days-
is involved in the regulation of sexual behavior and food appetite old rats started an intermittent-access (IA) ethanol drinking protocol
through its agonist activity on MC3R and MC4R melanocortin [63]: the animals were given access to 20% ethanol during three 24-
receptors (MC3Rs and MC4Rs) expressed in the hippocampus, hour-sessions per week, for 4 weeks. It has been reported that 4
paraventricular nucleus of the hypothalamus (PVN), ventral teg- weeks is enough time for the animals to reach their maximum level
mental area (VTA) and nucleus accumbens (NAc) [33]. Of note, α- of consumption through the IA protocol [63]. Drinking sessions
MSH has also been implicated in the modulation of ethanol intake started at the beginning of the dark cycle. Each ethanol-naive rat
[34]. The administration of a nonspecific agonist (melanotan-II) of was given access to one bottle of 20% v⁄v ethanol and one bottle of
both MC3Rs and MC4Rs reduces voluntary ethanol consumption in water. After 24 hours, the ethanol bottle was replaced with a second
C57BL/6J mice [35] and in selectively-bred alcohol-preferring rats water bottle that was available for the next 24 hours. The placement
[36]. However, when this agonist is administered to MC4R knock- of the ethanol bottle was alternated each ethanol drinking session to
out mice no effect was observed, revealing that MC4Rs (and not avoid for side preference. The control group drank only water ad
MC3Rs) are responsible for reducing alcohol intake [37]. Consis- libitum. Ethanol and water intakes were recorded daily. As rat
tent with this idea, the administration of a selective activator of strains that were not selectively bred for high preference for ethanol
MC4Rs at the NAc and VTA diminished voluntary alcohol con- are not homogeneous in the amount of ethanol ingested (such as
sumption in rats [38]. Moreover, evidence in rats indicates that Long-Evans, Wistar and Sprague-Dawley) [63, 64], after the 4
chronic ethanol consumption reduces α-MSH levels in the hypo- weeks acquisition period we selected the animals that drank more
thalamus [30, 39, 40, 41], while an increase in α-MSH is observed than 2 g ethanol/kg/day to carry out our studies (referred as ethanol-
in acute ethanol-treated rats [42]. Nevertheless, the effect of alcohol preferring rats) [65]. Ethanol-preferring rats were separated ran-
consumption on the expression of MC4Rs in the brain has not yet domly in 2 groups (n = 7 per group) and in the last 2 days the IA
been studied. protocol one group received a daily i.p. dose of 180 µg/kg of a spe-
cific MC4R peptide-agonist (Cyclo(Β-Ala-His-D-Phe-Arg-Trp-
While chronic ethanol treatment significantly attenuates α-MSH Glu)-NH2, Phoenix Pharmaceuticals, CA), while the other group
level in the pituitary, arcuate nucleus (ARC) and substantia nigra was injected with saline. The same was performed in parallel in the
(Rainero et al. 1990), the use of acute protocols elevate the hypo- respective two water-drinking control groups; (n = 7 per group).
thalamic POMC mRNA expression (Rasmussen et al. 1998). We selected this dose based on the work of Liu et al. [66] who ad-
The MC system has been linked for many years with anti- ministered i.p. the MC4R agonist peptide (Butir-His-D-Phe-Arg-
inflammatory effects in the brain [15, 43]. Interestingly, the MC4R Trp-Sar-NH2) in a similar way to that used in this study. These
Melanocortin-4 Receptor Activation Reduces Ethanol-induced Neuroinflammation Current Pharmaceutical Design, 2019, Vol. 25, No. 00 3

authors reported that increased expression of IL-1β and TNF-α


evoked by a model of brain damage in rats was strongly counter-
acted with this dose of the agonist. Same doses also decreased the
expression of TNF-α when administered i.p. in a model of cerebral
ischemia in gerbils [52].
All animal procedures are in accordance with the Guide for the
Care and Use of Laboratory Animals of the National Institutes of
Health and were approved by the Bioethical and Biosafety Commit-
tee of Universidad Autónoma de Chile (Ethical Approval Record
BE003-14, 2016).

2.2. Western Blot and Enzyme-linked Immunosorbent Assay


One day after the administration of the MC4R agonist, the ani-
mals were anesthetized with ketamine/xylazine (10:1 mg/kg of
body weight, i.p.), perfused with saline, decapitated and the brains
removed. Hippocampus, hypothalamus and prefrontal cortex tissues
were extracted and homogenized with an Ultra-Turrax homogenizer
in buffer containing Tris-HCl 100 mM pH 7.4, EDTA 5 mM, SDS
1%, PMSF 1 µM and a protease inhibitor cocktail (Pierce,
Rockford, IL, USA) (1 ml lysis buffer per 0.1 g tissue). Then, the
samples were centrifuged at 14.000 x g for 10 min at 4°C to remove Fig. (1). Ethanol intake in ethanol-preferring and non-preferring rats at
tissue debris. Protein concentration was determined in the super- the beginning and end of the 4-week IA protocol. After the first day of
natants with the Bio-Rad protein assay kit (Bio-Rad, Richmond, access to ethanol, animals were separated into 2 groups: ethanol-non -
CA, USA). IL-6, IL-1β and TNF-α levels were determined in the preferring rats if they drank a maximum of 2 g/kg/24 h (No-P first session)
hippocampus, hypothalamus and prefrontal cortex homogenates by and ethanol-preferring rats if they drank more than 2 g/kg/24 h (P first ses-
ELISA (IL-6, IL-1β and TNF-α EIA kit, Enzo Life Science, USA) sion). Then, the voluntary consumption of alcohol was recorded in the last
according to the manufacturer’s protocol. For the assay, 100 µL of day of the 4-week IA protocol (No-P final session and P final session).
sample was added per ELISA plate well. The results were normal- ANOVA *** p < 0.001; * p < 0.05. (A higher resolution / colour version of
ized by protein amount to pg/g tissue. this figure is available in the electronic copy of the article).
Hippocampus and hypothalamus homogenates were also ana-
lyzed by Western blot with a MC4R primary antibody (diluted In the last 2 days of the 4-week ethanol-acquisition stage, an
1:500) (sc-55567, Santa Cruz Biotechnology, Inc.). As loading MC4R-agonist synthetic peptide was administered (180 µg/kg daily
control, β-tubulin levels were also determined in the same blots i.p. doses) to one group of ethanol-preferring rats and to one group
(diluted 1:2000 primary antibody sc-9104, Santa Cruz Biotechnol- of water-drinking control rats. Then, the levels of the proinflamma-
ogy, Inc.). After the incubation with the corresponding HRP- tory cytokines IL-6, IL-1β and TNF-α were determined in the hip-
conjugated secondary antibodies, blotting membranes were re- pocampus, hypothalamus and prefrontal cortex homogenates by
vealed for chemiluminescence with Pierce ECL Western Blotting ELISA. We chose these proinflammatory cytokines because they
Substrate. The bands were quantified by densitometry with ImageJ are the ones that undergo major changes in the brain in response to
software. alcohol consumption [25]. As can be seen in Fig. 2, alcohol con-
3. RESULTS AND DISCUSSION sumption markedly increased the levels of these cytokines in the
three studied brain areas when compared to the rats that drank only
After 4 weeks of acquiring the preference for ethanol intake, the water (IL-1β: 12-fold, 22-fold and 14-fold; TNF-α: 5-fold, 4-fold
rats showed high heterogeneity in their consumption levels (Fig. 1), and 4-fold; IL-6: 7-fold, 6-fold and 4-fold in hippocampus, hypo-
most likely due to idiosyncratic heterogeneity of ethanol taste per- thalamus and prefrontal cortex, respectively). Although the increase
ception. Clearly, there were ethanol-preferring animals that initially of these pro-inflammatory cytokines may suggest their implication
showed a high alcohol intake (3.66 ± 0.27 g/k/day), and this con- in the alcohol-induced neuroinflammation, we cannot rule out other
sumption was increasing as the IA protocol progressed until reach- inflammatory mediators, including inflammatory lipids such as
ing an intake of 5.65 ± 0.41 g/k/day. These results are in agreement prostaglandins, whose synthesis is stimulated by ethanol consump-
with previous evidence reported for different strains of out-bred tion [68].
rats, which also exhibit an increase in their alcohol consumption
when subjected to the IA protocol [65]. This response could repre- Interestingly, the administration of the MC4R agonist in the last
sent both a mechanism of reinforcement and addiction to ethanol, as 2 days of the IA protocol decreased the levels of the three cytokines
well as a better perception of the taste of ethanol as chronic con- to than water drinking animals. These results demonstrate that
sumption progresses. Tang et al. [67] demonstrated that consump- MC4R activation inhibits the neuroinflammatory response induced
tion of ethanol augments its oral acceptability in adolescent rats by by heavy alcohol consumption, reported in models of neuroinflam-
a mechanism mediated, at least in part, by a decrease in the respon- mation produced by other mechanisms (e.g. LPS-induced neuroin-
siveness of the peripheral taste system to ethanol per se, rather than flammation [48, 49, 50], cerebral ischemia [51, 52], kainic acid-
to its bitter and sweet flavor components. induced excitotoxic damage [53] and spinal cord injury [54, 55]).
Although not significant, the administration of the agonist also
In contrast, in the group of rats selected as non-preferring the changed cytokine levels in the non-ethanol exposed animals. How-
initial average consumption was 2.02 ± 0.13 g/k/day and, unlike the ever, these alterations were not consistent between the different
preferring rats, the consumption of alcohol in this group decreased cytokines or the different brain areas analyzed. In fact, in some
as they progressed in the IA protocol (1.2 ± 0.24 g/k/day). To our cases the cytokine levels increase, in others, the opposite was found
knowledge, this effect of decreasing the voluntary consumption of or remain unchanged. Therefore, we believe that these inconsistent
alcohol in non-preferring out-bred rats subjected to the IA protocol alterations likely denote the inherent variability of the technique of
has not been previously reported, and probably reflects the genera- detection. Of note, the weight gain of the animals at the end of the
tion of a certain degree of aversion to ethanol intake. protocol showed no differences between the different experimental
groups (data not shown).
4 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Flores-Bastías et al.

Fig. (2). Levels of proinflammatory cytokines in rats that consumed alcohol, and effect of a synthetic MC4R-agonist peptide. Ethanol-preferring rats
were separated randomly in 2 groups (n = 7 per group) and in the last 2 days the IA protocol one group received a daily i.p. dose of 180 µg/kg of a specific
MC4R peptide-agonist (EtOH+Ago), while the other group was injected with saline (EtOH). The same procedure was performed in the respective two water-
drinking control groups (Ctrl+Ago and Ctrl) (n = 7 per group). One day after the administration of the MC4R agonist or saline, hippocampus, hypothalamus
and prefrontal cortex tissues were extracted, homogenized, and IL-6, IL-1β and TNF-α levels were determined by ELISA. ANOVA *** p < 0.001; ** p<0.01.
(A higher resolution / colour version of this figure is available in the electronic copy of the article).

Fig. (3). Expression of MC4R in hippocampus and hypothalamus of rats that consumed alcohol. At the end of the IA protocol, MC4R levels were deter-
mined by western blot in hippocampus (A) and hypothalamus (B) of water-drinking (Ctrl) and ethanol-drinking (EtOH) rats, that were not treated with MC4R
agonist (n = 7 per group). β-tubulin levels were determined as loading control. Three representative lanes of the western blots from each group are shown. Bars
represent MC4R/β-tubulin ratio. T-test ** p < 0.01. (A higher resolution / colour version of this figure is available in the electronic copy of the article).

Due to the marked effect of the MC4R agonist on the alcohol tions due to ethanol consumption, such as the hippocampus and the
intake-induced neuroinflammation, we examined if there were hypothalamus. As seen in Fig. 3, western blot analysis shows that
changes in the expression of MC4R in brain areas that suffer altera- alcohol consumption increases MC4R expression in both hippo-
Melanocortin-4 Receptor Activation Reduces Ethanol-induced Neuroinflammation Current Pharmaceutical Design, 2019, Vol. 25, No. 00 5

campus and hypothalamus. This finding is novel since although it [3] Nutt DJ, King LA, Phillips LD. Drug harms in the UK: a multicrite-
was reported that α-MSH levels are decreased in the hippocampus ria decision analysis. Lancet 2010; 376(9752): 1558-65.
of rats that drink alcohol [30, 39, 40, 41], reports that account for an [http://dx.doi.org/10.1016/S0140-6736(10)61462-6] [PMID:
alteration in the expression of MC4R are missing. It is possible that 21036393]
this increase in MC4R expression reflects a brain compensatory [4] Baan R, Straif K, Grosse Y, et al. Carcinogenicity of alcoholic
beverages. Lancet Oncol 2007; 8(4): 292-3.
mechanism due to the decrease in α-MSH levels by chronic alcohol
[http://dx.doi.org/10.1016/S1470-2045(07)70099-2] [PMID:
intake or even a defense response to the inflammatory process trig- 17431955]
gered by alcohol consumption. Because MC4R signaling activates [5] Shield KD, Parry C, Rehm J. Chronic diseases and conditions re-
the CREB/BDNF pathway [69] which is fundamental for neuronal lated to alcohol use. Alcohol Res 2013; 35(2): 155-73.
plasticity and cognitive processes, we could speculate that increased [PMID: 24881324]
MC4R expression could have positive effects on memory and learn- [6] Crews FT, Nixon K. Mechanisms of neurodegeneration and regen-
ing. In addition, considering that BDNF has been described as hav- eration in alcoholism. Alcohol Alcohol 2009; 44(2): 115-27.
ing anti-inflammatory actions in the brain [70], it is likely that not [http://dx.doi.org/10.1093/alcalc/agn079] [PMID: 18940959]
only could improve cognitive processes, but it may also decrease [7] Peterson JB, Rothfleisch J, Zelazo PD, Pihl RO. Acute alcohol
the neuro-glial inflammation evoked by ethanol consumption. intoxication and cognitive functioning. J Stud Alcohol 1990; 51(2):
114-22.
CONCLUSION [http://dx.doi.org/10.15288/jsa.1990.51.114] [PMID: 2308348]
[8] Whiteford HA, Degenhardt L, Rehm J, et al. Global burden of
In this study, we found that the i.p. administration of a synthetic disease attributable to mental and substance use disorders: findings
MC4R agonist prevents neuroinflammation induced by alcohol from the Global Burden of Disease Study 2010. Lancet 2013;
consumption in the hippocampus, hypothalamus and prefrontal 382(9904): 1575-86.
cortex. In addition, high alcohol consumption produced an increase [http://dx.doi.org/10.1016/S0140-6736(13)61611-6] [PMID:
in MC4R expression in the hippocampus and hypothalamus. These 23993280]
results could explain the effect of α-MSH and synthetic MC4R [9] Kushner MG, Abrams K, Borchardt C. The relationship between
agonists in decreasing alcohol intake, through the reduction of the anxiety disorders and alcohol use disorders: a review of major per-
ethanol-induced inflammatory response in the brain. spectives and findings. Clin Psychol Rev 2000; 20(2): 149-71.
[http://dx.doi.org/10.1016/S0272-7358(99)00027-6] [PMID:
ETHICS APPROVAL AND CONSENT TO PARTICIPATE 10721495]
[10] Rose ME, Grant JE. Alcohol-induced blackout. Phenomenology,
Not applicable. biological basis, and gender differences. J Addict Med 2010; 4(2):
61-73.
HUMAN AND ANIMAL RIGHTS [http://dx.doi.org/10.1097/ADM.0b013e3181e1299d] [PMID:
No Animals/Humans were used for studies that are the basis of 21769024]
this research. [11] Israel Y, Karahanian E, Ezquer F, et al. Acquisition, Maintenance
and Relapse-Like Alcohol Drinking: Lessons from the UChB Rat
CONSENT FOR PUBLICATION Line. Front Behav Neurosci 2017; 11: 57.
[http://dx.doi.org/10.3389/fnbeh.2017.00057] [PMID: 28420969]
Not applicable.
[12] Coller JK, Hutchinson MR. Implications of central immune signal-
AVAILABILITY OF DATA AND MATERIALS   ing caused by drugs of abuse: mechanisms, mediators and new
therapeutic approaches for prediction and treatment of drug de-
Not applicable. pendence. Pharmacol Ther 2012; 134(2): 219-45.
[http://dx.doi.org/10.1016/j.pharmthera.2012.01.008] [PMID:
22316499]
FUNDING   [13] Flores-Bastías O, Karahanian E. Neuroinflammation produced by
This work was supported by Programa de Investigación heavy alcohol intake is due to loops of interactions between Toll-
Asociativa (PIA-CONICYT) Anillo de Ciencia y Tecnología like 4 and TNF receptors, peroxisome proliferator-activated recep-
ACT1411 (to EK and JAO), DIUA149-2019 (to EK) and tors and the central melanocortin system: A novel hypothesis and
new therapeutic avenues. Neuropharmacology 2018; 128: 401-7.
FONDECYT 1160710 (to JAO).
[http://dx.doi.org/10.1016/j.neuropharm.2017.11.003] [PMID:
CONFLICT OF INTEREST 29113896]
[14] Crews FT, Zou J, Qin L. Induction of innate immune genes in brain
The authors declare no conflict of interest, financial or create the neurobiology of addiction. Brain Behav Immun 2011;
other-wise. 25(Suppl. 1): S4-S12.
[http://dx.doi.org/10.1016/j.bbi.2011.03.003] [PMID: 21402143]
ACKNOWLEDGEMENTS [15] Orellana JA, Cerpa W, Carvajal MF, et al. New implications for the
melanocortin system in alcohol drinking behavior in adolescents:
Declared none. The glial dysfunction hypothesis. Front Cell Neurosci 2017; 11: 90.
[http://dx.doi.org/10.3389/fncel.2017.00090] [PMID: 28424592]
[16] Blanco AM, Guerri C. Ethanol intake enhances inflammatory me-
REFERENCES diators in brain: role of glial cells and TLR4/IL-1RI receptors. Front
Biosci 2007; 12: 2616-30.
[1] World Health Organization. Global Status Report on Alcohol and
[http://dx.doi.org/10.2741/2259] [PMID: 17127267]
Health 2014. Available from: www.who.int/substance_abuse/ pub-
[17] Walker DW, Barnes DE, Zornetzer SF, Hunter BE, Kubanis P.
lications/global_alcohol_report/en/
Neuronal loss in hippocampus induced by prolonged ethanol con-
[2] Lim SS, Vos T, Flaxman AD, et al. A comparative risk assessment
sumption in rats. Science 1980; 209(4457): 711-3.
of burden of disease and injury attributable to 67 risk factors and
[http://dx.doi.org/10.1126/science.7394532] [PMID: 7394532]
risk factor clusters in 21 regions, 1990-2010: a systematic analysis
[18] Franke H, Kittner H, Berger P, Wirkner K, Schramek J. The reac-
for the Global Burden of Disease Study 2010. Lancet 2012;
tion of astrocytes and neurons in the hippocampus of adult rats dur-
380(9859): 2224-60.
ing chronic ethanol treatment and correlations to behavioral im-
[http://dx.doi.org/10.1016/S0140-6736(12)61766-8] [PMID:
pairments. Alcohol 1997; 14(5): 445-54.
23245609]
[http://dx.doi.org/10.1016/S0741-8329(96)00209-1] [PMID:
9305459]
6 Current Pharmaceutical Design, 2019, Vol. 25, No. 00 Flores-Bastías et al.

[19] He J, Crews FT. Increased MCP-1 and microglia in various regions [http://dx.doi.org/10.1007/978-1-4419-6354-3_3] [PMID:
of the human alcoholic brain. Exp Neurol 2008; 210(2): 349-58. 21222258]
[http://dx.doi.org/10.1016/j.expneurol.2007.11.017] [PMID: [34] Olney JJ, Navarro M, Thiele TE. Targeting central melanocortin
18190912] receptors: a promising novel approach for treating alcohol abuse
[20] Ward RJ, Colivicchi MA, Allen R, et al. Neuro-inflammation in- disorders. Front Neurosci 2014; 8: 128.
duced in the hippocampus of ‘binge drinking’ rats may be mediated [http://dx.doi.org/10.3389/fnins.2014.00128] [PMID: 24917782]
by elevated extracellular glutamate content. J Neurochem 2009; [35] Navarro M, Cubero I, Knapp DJ, Thiele TE. MTII-induced reduc-
111(5): 1119-28. tion of voluntary ethanol drinking is blocked by pretreatment with
[http://dx.doi.org/10.1111/j.1471-4159.2009.06389.x] [PMID: AgRP-(83-132). Neuropeptides 2003; 37(6): 338-44.
19765190] [http://dx.doi.org/10.1016/j.npep.2003.10.003] [PMID: 14698676]
[21] Zou J, Crews F. Induction of innate immune gene expression cas- [36] Ploj K, Roman E, Kask A, et al. Effects of melanocortin receptor
cades in brain slice cultures by ethanol: key role of NF-κB and ligands on ethanol intake and opioid peptide levels in alcohol-
proinflammatory cytokines. Alcohol Clin Exp Res 2010; 34(5): preferring AA rats. Brain Res Bull 2002; 59(2): 97-104.
777-89. [http://dx.doi.org/10.1016/S0361-9230(02)00844-4] [PMID:
[http://dx.doi.org/10.1111/j.1530-0277.2010.01150.x] [PMID: 12379439]
20201932] [37] Navarro M, Lerma-Cabrera JM, Carvajal F, Lowery EG, Cubero I,
[22] Chandel NS, Trzyna WC, McClintock DS, Schumacker PT. Role of Thiele TE. Assessment of voluntary ethanol consumption and the
oxidants in NF-kappa B activation and TNF-alpha gene transcrip- effects of a melanocortin (MC) receptor agonist on ethanol intake in
tion induced by hypoxia and endotoxin. J Immunol 2000; 165(2): mutant C57BL/6J mice lacking the MC-4 receptor. Alcohol Clin
1013-21. Exp Res 2011; 35(6): 1058-66.
[http://dx.doi.org/10.4049/jimmunol.165.2.1013] [PMID: [http://dx.doi.org/10.1111/j.1530-0277.2011.01438.x] [PMID:
10878378] 21332528]
[23] Lieber CS. Microsomal ethanol-oxidizing system (MEOS): the first [38] Lerma-Cabrera JM, Carvajal F, de la Torre L, et al. Control of food
30 years (1968-1998)--a review. Alcohol Clin Exp Res 1999; 23(6): intake by MC4-R signaling in the lateral hypothalamus, nucleus ac-
991-1007. cumbens shell and ventral tegmental area: interactions with ethanol.
[http://dx.doi.org/10.1097/00000374-199906000-00006] [PMID: Behav Brain Res 2012; 234(1): 51-60.
10397283] [http://dx.doi.org/10.1016/j.bbr.2012.06.006] [PMID: 22713514]
[24] Cao Q, Mak KM, Lieber CS. Cytochrome P4502E1 primes macro- [39] Rainero I, De Gennaro T, Visentin G, et al. Effects of chronic etha-
phages to increase TNF-alpha production in response to lipopoly- nol treatment on alpha-MSH concentrations in rat brain and pitui-
saccharide. Am J Physiol Gastrointest Liver Physiol 2005; 289(1): tary. Neuropeptides 1990; 15(3): 139-41.
G95-G107. [http://dx.doi.org/10.1016/0143-4179(90)90145-O] [PMID:
[http://dx.doi.org/10.1152/ajpgi.00383.2004] [PMID: 15961886] 2174518]
[25] Crews FT, Sarkar DK, Qin L, Zou J, Boyadjieva N, Vetreno RP. [40] Lerma-Cabrera JM, Carvajal F, Alcaraz-Iborra M, et al. Adolescent
Neuroimmune function and the consequences of alcohol exposure. binge-like ethanol exposure reduces basal α-MSH expression in the
Alcohol Res 2015; 37(2): 331-341, 344-351. hypothalamus and the amygdala of adult rats. Pharmacol Biochem
[PMID: 26695754] Behav 2013; 110: 66-74.
[26] Barson JR, Leibowitz SF. Hypothalamic neuropeptide signaling in [http://dx.doi.org/10.1016/j.pbb.2013.06.006] [PMID: 23792540]
alcohol addiction. Prog Neuropsychopharmacol Biol Psychiatry [41] Sprow GM, Rinker JA, Lowery-Gointa EG, Sparrow AM, Navarro
2016; 65: 321-9. M, Thiele TE. Lateral hypothalamic melanocortin receptor signal-
[http://dx.doi.org/10.1016/j.pnpbp.2015.02.006] [PMID: 25689818] ing modulates binge-like ethanol drinking in C57BL/6J mice. Ad-
[27] Rada P, Avena NM, Leibowitz SF, Hoebel BG. Ethanol intake is dict Biol 2016; 21(4): 835-46.
increased by injection of galanin in the paraventricular nucleus and [http://dx.doi.org/10.1111/adb.12264] [PMID: 25975524]
reduced by a galanin antagonist. Alcohol 2004; 33(2): 91-7. [42] Shelkar GP, Kale AD, Singh U, Singru PS, Subhedar NK, Kokare
[http://dx.doi.org/10.1016/S0741-8329(04)00097-7] [PMID: DM. Alpha-melanocyte stimulating hormone modulates ethanol
15528006] self-administration in posterior ventral tegmental area through
[28] Schneider ER, Rada P, Darby RD, Leibowitz SF, Hoebel BG. melanocortin-4 receptors. Addict Biol 2015; 20(2): 302-15.
Orexigenic peptides and alcohol intake: differential effects of [http://dx.doi.org/10.1111/adb.12126] [PMID: 24635847]
orexin, galanin, and ghrelin. Alcohol Clin Exp Res 2007; 31(11): [43] Macaluso A, McCoy D, Ceriani G, et al. Antiinflammatory influ-
1858-65. ences of alpha-MSH molecules: central neurogenic and peripheral
[http://dx.doi.org/10.1111/j.1530-0277.2007.00510.x] [PMID: actions. J Neurosci 1994; 14(4): 2377-82.
17850217] [http://dx.doi.org/10.1523/JNEUROSCI.14-04-02377.1994]
[29] Barson JR, Carr AJ, Soun JE, et al. Opioids in the hypothalamic [PMID: 8158274]
paraventricular nucleus stimulate ethanol intake. Alcohol Clin Exp [44] Benjamins JA, Nedelkoska L, Bealmear B, Lisak RP. ACTH pro-
Res 2010; 34(2): 214-22. tects mature oligodendroglia from excitotoxic and inflammation-
[http://dx.doi.org/10.1111/j.1530-0277.2009.01084.x] [PMID: related damage in vitro. Glia 2013; 61(8): 1206-17.
19951300] [http://dx.doi.org/10.1002/glia.22504] [PMID: 23832579]
[30] Navarro M, Cubero I, Knapp DJ, Breese GR, Thiele TE. Decreased [45] Caruso C, Durand D, Schiöth HB, Rey R, Seilicovich A, Lasaga M.
immunoreactivity of the melanocortin neuropeptide alpha- Activation of melanocortin 4 receptors reduces the inflammatory
melanocyte-stimulating hormone (alpha-MSH) after chronic etha- response and prevents apoptosis induced by lipopolysaccharide and
nol exposure in Sprague-Dawley rats. Alcohol Clin Exp Res 2008; interferon-gamma in astrocytes. Endocrinology 2007; 148(10):
32(2): 266-76. 4918-26.
[http://dx.doi.org/10.1111/j.1530-0277.2007.00578.x] [PMID: [http://dx.doi.org/10.1210/en.2007-0366] [PMID: 17595227]
18162070] [46] Selkirk JV, Nottebaum LM, Lee J, Yang W, Foster AC, Lechner
[31] Thorsell A, Slawecki CJ, Ehlers CL. Effects of neuropeptide Y and SM. Identification of differential melanocortin 4 receptor agonist
corticotropin-releasing factor on ethanol intake in Wistar rats: inter- profiles at natively expressed receptors in rat cortical astrocytes and
action with chronic ethanol exposure. Behav Brain Res 2005; recombinantly expressed receptors in human embryonic kidney
161(1): 133-40. cells. Neuropharmacology 2007; 52(2): 459-66.
[http://dx.doi.org/10.1016/j.bbr.2005.01.016] [PMID: 15904720] [http://dx.doi.org/10.1016/j.neuropharm.2006.08.015] [PMID:
[32] Hadley ME, Haskell-Luevano C. The proopiomelanocortin system. 17095023]
Ann N Y Acad Sci 1999; 885: 1-21. [47] Caruso C, Mohn C, Karara AL, et al. α-melanocyte-stimulating
[http://dx.doi.org/10.1111/j.1749-6632.1999.tb08662.x] [PMID: hormone through melanocortin-4 receptor inhibits nitric oxide syn-
10816638] thase and cyclooxygenase expression in the hypothalamus of male
[33] Mountjoy KG. Distribution and function of melanocortin receptors rats. Neuroendocrinology 2004; 79(5): 278-86.
within the brain. Adv Exp Med Biol 2010; 681: 29-48. [http://dx.doi.org/10.1159/000079321] [PMID: 15218320]
Melanocortin-4 Receptor Activation Reduces Ethanol-induced Neuroinflammation Current Pharmaceutical Design, 2019, Vol. 25, No. 00 7

[48] Ichiyama T, Zhao H, Catania A, Furukawa S, Lipton JM. α- [http://dx.doi.org/10.1159/000097313] [PMID: 9326743]
melanocyte-stimulating hormone inhibits NF-kappaB activation and [60] Muceniece R, Zvejniece L, Liepinsh E, et al. The MC3 receptor
IkappaBalpha degradation in human glioma cells and in experimen- binding affinity of melanocortins correlates with the nitric oxide
tal brain inflammation. Exp Neurol 1999; 157(2): 359-65. production inhibition in mice brain inflammation model. Peptides
[http://dx.doi.org/10.1006/exnr.1999.7064] [PMID: 10364447] 2006; 27(6): 1443-50.
[49] Ichiyama T, Sakai T, Catania A, Barsh GS, Furukawa S, Lipton JM. [http://dx.doi.org/10.1016/j.peptides.2005.12.002] [PMID:
Systemically administered alpha-melanocyte-stimulating peptides 16414147]
inhibit NF-kappaB activation in experimental brain inflammation. [61] Galimberti D, Baron P, Meda L, et al. Alpha-MSH peptides inhibit
Brain Res 1999; 836(1-2): 31-7. production of nitric oxide and tumor necrosis factor-alpha by mi-
[http://dx.doi.org/10.1016/S0006-8993(99)01584-X] [PMID: croglial cells activated with beta-amyloid and interferon gamma.
10415402] Biochem Biophys Res Commun 1999; 263(1): 251-6.
[50] Muceniece R, Zvejniece L, Kirjanova O, et al. β-MSH inhibits [http://dx.doi.org/10.1006/bbrc.1999.1276] [PMID: 10486285]
brain inflammation via MC(3)/(4) receptors and impaired NF- [62] Carniglia L, Durand D, Caruso C, Lasaga M. Effect of NDP-α-
kappaB signaling. J Neuroimmunol 2005; 169(1-2): 13-9. MSH on PPAR-γ and -β expression and anti-inflammatory cytokine
[http://dx.doi.org/10.1016/j.jneuroim.2005.07.024] [PMID: release in rat astrocytes and microglia. PLoS One 2013; 8(2)e57313
16154641] [http://dx.doi.org/10.1371/journal.pone.0057313] [PMID:
[51] Giuliani D, Mioni C, Altavilla D, et al. Both early and delayed 23468969]
treatment with melanocortin 4 receptor-stimulating melanocortins [63] Simms JA, Steensland P, Medina B, et al. Intermittent access to
produces neuroprotection in cerebral ischemia. Endocrinology 20% ethanol induces high ethanol consumption in Long-Evans and
2006; 147(3): 1126-35. Wistar rats. Alcohol Clin Exp Res 2008; 32(10): 1816-23.
[http://dx.doi.org/10.1210/en.2005-0692] [PMID: 16254026] [http://dx.doi.org/10.1111/j.1530-0277.2008.00753.x] [PMID:
[52] Spaccapelo L, Bitto A, Galantucci M, et al. Melanocortin MC₄ 18671810]
receptor agonists counteract late inflammatory and apoptotic re- [64] Moorman DE, Aston-Jones G. Orexin-1 receptor antagonism de-
sponses and improve neuronal functionality after cerebral ischemia. creases ethanol consumption and preference selectively in high-
Eur J Pharmacol 2011; 670(2-3): 479-86. ethanol--preferring Sprague--Dawley rats. Alcohol 2009; 43(5):
[http://dx.doi.org/10.1016/j.ejphar.2011.09.015] [PMID: 21946115] 379-86.
[53] Forslin Aronsson A, Spulber S, Oprica M, Winblad B, Post C, [http://dx.doi.org/10.1016/j.alcohol.2009.07.002] [PMID:
Schultzberg M. α-MSH rescues neurons from excitotoxic cell death. 19671464]
J Mol Neurosci 2007; 33(3): 239-51. [65] Carnicella S, Ron D, Barak S. Intermittent ethanol access schedule
[http://dx.doi.org/10.1007/s12031-007-0019-2] [PMID: 17952633] in rats as a preclinical model of alcohol abuse. Alcohol 2014; 48(3):
[54] van de Meent H, Hamers FP, Lankhorst AJ, Joosten EA, Gispen 243-52.
WH. Beneficial effects of the melanocortin alpha-melanocyte- [http://dx.doi.org/10.1016/j.alcohol.2014.01.006] [PMID:
stimulating hormone on clinical and neurophysiological recovery 24721195]
after experimental spinal cord injury. Neurosurgery 1997; 40(1): [66] Liu D, Zhang HG, Zhao ZA, et al. Melanocortin MC4 receptor
122-30. agonists alleviate brain damage in abdominal compartment syn-
[PMID: 8971834] drome in the rat. Neuropeptides 2015; 49: 55-61.
[55] Lankhorst AJ, Duis SE, ter Laak MP, Joosten EA, Hamers FP, [http://dx.doi.org/10.1016/j.npep.2014.12.003] [PMID: 25616531]
Gispen WH. Functional recovery after central infusion of alpha- [67] Tang J, Youngentob SL, Glendinning JI. Postnatal Exposure to
melanocyte-stimulating hormone in rats with spinal cord contusion ethanol increases its oral acceptability to adolescent rats. Chem
injury. J Neurotrauma 1999; 16(4): 323-31. Senses 2018; 43(8): 655-64.
[http://dx.doi.org/10.1089/neu.1999.16.323] [PMID: 10225218] [http://dx.doi.org/10.1093/chemse/bjy056] [PMID: 30169758]
[56] Catania A. Neuroprotective actions of melanocortins: a therapeutic [68] Horrobin DF. Essential fatty acids, prostaglandins, and alcoholism:
opportunity. Trends Neurosci 2008; 31(7): 353-60. an overview. Alcohol Clin Exp Res 1987; 11(1): 2-9.
[http://dx.doi.org/10.1016/j.tins.2008.04.002] [PMID: 18550183] [http://dx.doi.org/10.1111/j.1530-0277.1987.tb01250.x] [PMID:
[57] Delgado R, Carlin A, Airaghi L, et al. Melanocortin peptides inhibit 3032012]
production of proinflammatory cytokines and nitric oxide by acti- [69] Caruso C, Carniglia L, Durand D, Gonzalez PV, Scimonelli TN,
vated microglia. J Leukoc Biol 1998; 63(6): 740-5. Lasaga M. Melanocortin 4 receptor activation induces brain-derived
[http://dx.doi.org/10.1002/jlb.63.6.740] [PMID: 9620667] neurotrophic factor expression in rat astrocytes through cyclic
[58] Rajora N, Boccoli G, Burns D, Sharma S, Catania AP, Lipton JM. AMP-protein kinase A pathway. Mol Cell Endocrinol 2012; 348(1):
α-MSH modulates local and circulating tumor necrosis factor-α in 47-54.
experimental brain inflammation. J Neurosci 1997; 17(6): 2181-6. [http://dx.doi.org/10.1016/j.mce.2011.07.036] [PMID: 21803120]
[http://dx.doi.org/10.1523/JNEUROSCI.17-06-02181.1997] [70] Xu D, Lian D, Wu J, et al. Brain-derived neurotrophic factor re-
[PMID: 9045742] duces inflammation and hippocampal apoptosis in experimental
[59] Wong KY, Rajora N, Boccoli G, Catania A, Lipton JM. A potential Streptococcus pneumoniae meningitis. J Neuroinflammation 2017;
mechanism of local anti-inflammatory action of alpha-melanocyte- 14(1): 156.
stimulating hormone within the brain: modulation of tumor necrosis [http://dx.doi.org/10.1186/s12974-017-0930-6] [PMID: 28778220]
factor-alpha production by human astrocytic cells. Neuroimmuno-
modulation 1997; 4(1): 37-41.

You might also like