Melanina y Emocion

download Melanina y Emocion

of 7

Transcript of Melanina y Emocion

  • 7/28/2019 Melanina y Emocion

    1/7

    Peptides 25 (2004) 16051611

    Review

    Beyond skin color: emerging roles of melanin-concentrating hormonein energy homeostasis and other physiological functions

    Yuguang Shi

    Endocrine Research, Lilly Research Laboratories, DC 0545, Eli Lilly and Company, Lilly Corporate Center, Indianapo lis, IN 46285, USA

    Received 23 January 2004; accepted 24 February 2004

    Available online 11 September 2004

    Abstract

    Melanin-concentrating hormone (MCH) is a cyclic peptide that mediates its effects by the activation of two G-protein-coupled seven

    transmembrane receptors (MCHR1 and MCHR2) in humans. In contrast to its primary role in regulating skin color in fish, MCH has evolved

    in mammals to regulate dynamic physiological functions, from food intake and energy expenditure to behavior and emotion. Chronic infusion

    or transgenic expression of MCHstimulates feeding and increasesadipocity, whereas targeted deletion of MCH or its receptor (MCHR1) leads

    to resistance to diet-induced obesity with increased energy expenditure and thermogenesis. The involvement of MCH in energy homeostasis

    and in brain activity has also been validated in mice treated with non-peptide antagonists, suggesting that blockade of MCHR1 could provide

    a viable approach for treatment of obesity and certain neurological disorders. This review focuses on emerging roles of MCH in regulating

    central and peripheral mechanisms.

    Published by Elsevier Inc.

    Keywords: Melanin-concentrating hormone; Obesity; Agonist; Antagonist; Hyperphagia; Energy expenditure; Melanocyte-stimulating hormone; Anxiety

    Contents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1605

    2. MCH and pigmentation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1606

    3. MCH and energy homeostasis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1607

    4. MCH receptor as a drug target for obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1608

    5. MCH and social behaviors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1609

    6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1609

    Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1609

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1609

    1. Introduction

    Melanin-concentrating hormone (MCH) is a cyclic pep-

    tide originally isolated from the teleost fish as a pituitary

    hormone that regulates skin color [35,54]. In teleost fish,

    MCH causes aggregation of melanin granules thus lighten-

    Tel.: +1 317 276 6753; fax: +1 317 276 9574.

    E-mail address: shi [email protected].

    ing the skin, and its action is antagonized by -melanocyte

    stimulating hormone (-MSH) that causes melanin granule

    dispersion. The mammalian MCH is a 19-amino acid peptide

    that shares high homology with fish MCH, and is fully con-

    served in all mammals analyzed thus far [46,47,82]. MCH is

    generated via a proteolytic cleavage of the preprohormone of

    165 residues, generating two additional peptides, neuropep-

    tide E-I (NEI) and neuropeptide G-E (NGE). Two related

    peptides are also generated from the Pmch gene that en-

    0196-9781/$ see front matter. Published by Elsevier Inc.

    doi:10.1016/j.peptides.2004.02.023

  • 7/28/2019 Melanina y Emocion

    2/7

    1606 Y. Shi / Peptides 25 (2004) 16051611

    codes the MCH hormone. One is the MCH-gene-overprinted-

    polypeptide (MGOP peptide) produced by alternative frame

    reading to theMCH codingregion, whereas theother is a pep-

    tide named antisense RNA overlapping from MCH (AROM)

    generated by transcription of the DNA partially overlapping

    with the Pmch gene [13,21,52,78]. The function of those

    MCH related peptides remains to be identified.Biological function of MCH is mediated by two G-

    protein-coupled receptors known as MCHR1 and MCHR2.

    MCHR1 was simultaneously identified by several groups, us-

    ing different biochemical and pharmacological approaches,

    and the encoding gene was found to be identical to that of the

    previously cloned orphan receptor SLC-1 [4,19,38,63,64].

    Human MCHR1 is closely related to the somatostatin re-

    ceptor family (35% homology), and is highly homologous

    with MCHR1 from other mammalian species. MCHR2 was

    later identified by its limited sequence homology with a core

    region of MCHR1 [2,31,45,56,61,84]. In contrast to MCHR1

    that is conserved among all mammals, functional MCHR2 is

    only expressed in carnivores and primates, but not in rodents[77]. The multifunctional role of MCH is accomplished

    in part by the coupling of MCH receptors with different

    G-proteins. MCHR1 is primarily coupled to Gi and Gq, as

    evidenced by the fact that MCHR1 activation leads to

    suppression of forskolin-stimulated cAMP and reduction of

    intracellular Ca2+ [29]. In addition, MCHR1 activation was

    also found to stimulate other signal transduction pathways,

    including those mediated by protein kinase C, phospholipase

    C, and extracellular signal-regulated kinase [29,51]. In

    contrast, MCHR2 is believed to be exclusively coupled

    to Gq [2,31,61]. The two receptors share similar tissue

    distribution patterns within the CNS [68], although MCHR1is more abundant and exhibits a wider distribution pattern

    than MCHR2, whose function has yet to be determined due

    to its absence in rodents [77].

    Physiological function of MCH and its receptors was

    subjected to intensive investigation in recent years that was

    stimulated by an initial observation that over-expression of

    MCH was associated with the onset of obesity in ob/ob mice

    [53]. Cumulative evidence suggests that MCH contributes to

    a variety of physiological functions, from pigmentation and

    energy homeostasis to cognation and emotion (Fig. 1).

    2. MCH and pigmentation

    Although MCH was originally discovered in the pitu-

    itary of teleost fish for its regulation of skin melanocyte

    aggregation [35,54], its function in regulating skin color

    in higher mammals has not been confirmed. Coat color

    in mice is controlled by complex genetic traits [83]. The

    agouti protein is a major regulator of pigmentation in mice,

    which antagonizes the binding of melanocyte stimulating

    hormone (-MSH) to melanocortin 1 receptor (MC1R),

    switching melanin synthesis from eumelanin (black/brown)

    to pheomelanin (red/yellow). Dominant mutations in the

    Fig. 1. Schematic representation of MCH circuitry in the brain, and its pro-

    posed regulation of peripheral physiology. The arcuate nucleus (ARC) con-

    tains neurons that co-express-melanocyte-stimulating hormone (-MSH),

    an antagonist for MCH function, and cocaine and amphetamine-related tran-

    script (CART) that are activated when there is a positive energy balance or

    stimulated by leptin and insulin. The arcuate nucleus also contains neu-

    rons that are responsive to food restriction through the action of neuropep-

    tide Y (NPY) and agouti-related protein (AgRP). Neurons in the arcuate

    nucleus project to other brain nuclei that are involved in regulation of en-

    ergy balance, including lateral hypothalamus where melanocortin receptor-

    containing neurons that express orexigenic neuropeptide MCH and orexin

    are located, and paraventicular nucleus that contains neurons, which ex-

    press anorexigenic neuropeptides corticotropin-releasing factor (CRF) and

    thyroid-releasing hormone (TRH). Catabolic projections from the paraven-

    ticular nucleus and anabolic input from the lateral hypothalamus determine

    thenet neuronal output from thenucleus of thesolitarytract (NTS).Alterna-

    tively, MCH could also have direct local action as depicted by dashed lines.

    WAT, white adipose tissue; and BAT, brown adipose tissue.

    non-coding region of the mouse agouti gene results in

    yellow coat color, whereas ectopic expression in brain

    causes obesity and type II diabetes resulting from antago-

    nistic action of agouti on MC3 and MC4 receptors [55,83].

    Loss-of-function mutations at the MC1R are associated

    with a switch from eumelanin to pheomelanin production,

    resulting in a red or yellow coat color. In man, a number

    of loss-of-function mutations in MC1R are associated with

    red-hair and lightening of skin color, although activating

    mutations of the MC1R in man have not been reported [67].

    Like agouti in mice and agouti signal protein (ASIP) in hu-

    man, MCH also functionally antagonizes the action of MSH

    in fish and mammals, thus expecting some effects on coat and

    skin color. Yet, this has not been confirmed in animal models

    of MCH physiology. Mice, with targeted deletion of MCH or

    MCHR1, developed normal coat and skin color, suggesting

    that the MCH is not obligated to normal development of skin

  • 7/28/2019 Melanina y Emocion

    3/7

    Y. Shi / Peptides 25 (2004) 16051611 1607

    color in rodents. However, due to some differences in MCH

    function between rodents and humans, the possible regula-

    tory role of MCH on skin and hair color in humans cannot be

    ruled out. For example, MCH binding sites were identified in

    human SVK14 keratinocytes, and both MCH and its recep-

    tor are expressed and functional in human skin [18,32,65], as

    demonstrated by inhibition of the melanogenic actions of-MSH by MCH in cultured human melanocytes. Furthermore,

    autoantibodies against the MCHR1 were recently identified

    in patients with vitiligo, a common depigmentation disorder

    resulting from the destruction of functional melanocytes in

    the affected skin [25,37], although a causative role of the

    autoantibodies remains to be elucidated. Since MCH is al-

    most exclusively expressed in the brain, and melanocytes are

    mainly derived from the neural crest cells [85], it is not yet

    clear whether the MCHeffect on melanocytes is of peripheral

    or central origin (Fig. 1). Nevertheless, these findings neces-

    sitate further investigation to confirm a role of MCH on skin

    and hair pigmentation.

    3. MCH and energy homeostasis

    Although MCH was originally named as a melanin-

    concentrating hormone for its involvement in regulating skin

    color, it plays an even greater role in regulating physiological

    functions in the CNS of both fish [1,28,49] and higher mam-

    mals [34,47,50]. Cumulative evidence suggests that MCH

    plays an important role in maintaining energy homeostasis

    by regulating satiety, energy expenditure, and thermogenesis.

    The expression of both MCH and MCHR1 is up-regulated by

    genetic obesity and fooddeprivation [27,30,39,53]. Intracere-broventricular (ICV)injectionof MCHstimulatesfood intake

    in rats, which can be functionally antagonized by MSH, an

    inhibitor of appetite [39,58,79]. Chronic infusion of MCH or

    a peptide agonist induces hyperphagia and body weight gain

    [33], whereas continuous treatment of mice with a MCHR1

    peptide antagonist suppresses appetite and adipocity [72].

    Further evidence regarding a role of MCH in regulating

    energy homeostasis comes from animal models with genetic

    manipulation of MCH or MCHR1 expression. Transgenic

    expression of MCH leads to increased susceptibility to obe-

    sity and insulin resistance [40]. Targeted deletion of MCH

    and MCHR1 also supports a role for MCH in regulating en-

    ergy homeostasis. Mice lacking MCH are lean as a result

    of hypophagia and a slightly increased metabolic rate [73].

    Likewise, knockout mice with MCHR1 deficiency are resis-

    tant to obesity with increased energy expenditure [20,41].

    However, in comparison with hypophagia in MCH knock

    out mice, MCHR1-null mice present hyperphagia and hy-

    peractivity. Such differences are likely caused by deletion

    of MCH related peptides such as NEI, NGE, AROM, and

    MGOP [13,21,52,78], since MCHR1 has been demonstrated

    to be the only functional receptor for MCH-mediated activity

    in mice [77]. Consistent with a role for these peptides in con-

    tributing to the MCH knockout phenotype, NEI is reported

    to stimulate exploratory behavior, increase anxiety, and re-

    duce dopamine release [24]. NEI is also shown to antagonize

    the action of MCH on stress-induced release of adrenocorti-

    cotropin [11].

    As part of the complex central network involved in regula-

    tion of satiety and energy expenditure [5,16,22,66,70], MCH

    plays an important role in mediating the effects of leptin onenergy homeostasis [34,69]. As such, leptin deficiency in

    ob/ob mice was demonstrated to significantly increase MCH

    expression, whereas leptin administration leads to a dramatic

    decrease of MCH [53,60]. Ablation of MCH expression in

    ob/ob mice corrects the obese phenotype by increasing rest-

    ing energy expenditure and locomotive activity [71]. The

    data suggest that MCH functions downstream in the leptin-

    mediated signal transduction pathway. However, ablation of

    MCH expression only partially corrects the defective physio-

    logical effects of leptin deficiency, since the double-null mice

    still exhibit hyperphagia and hyperinsulinemia [71]. MCH is

    exclusively expressed in the lateral hypothalamus and zona

    incerta in close proximity with orexin containing neurons.MCH-expressing neurons project to nucleus of the solitary

    tract (NTS) (Fig. 1), parabrachial nucleus, and medial pre-

    frontal cortex regions, establishing connections throughout

    the brain [9,10,62]. Thus, alteration in MCH expression is ex-

    pected to change orexigenic peptide expression in the arcuate

    nucleus (ARC), such as neuropeptide Y (NPY) and agouti-

    related protein (AgRP), or orexin in the lateral hypothalamus.

    As a result of MCHdeficiency, POMC expression in the ARC

    is markedly diminished to counterbalance the diminution in

    satiety and body weight [71]. Such a compensatory change in

    POMC expression is not caused by lack of direct antagonism

    at the melanocortin receptors, as MCH and -MSH do notcompete with each other in receptor binding [39,57].

    In addition to a role in the central regulation of the feed-

    ing response, MCH also plays a role in regulating the levels

    of insulin, glucocorticoid, and thermogenesis (Fig. 1). Trans-

    genic over-expression and targeted deletion of MCH have

    reciprocal effects on glucocorticoid and insulin levels. In ad-

    dition to hyperinsulinemia, transgenic mice over-expressing

    MCH also developed islet cell hyperplasia, suggesting a se-

    vere insulin resistance. In supporting a role of MCH in reg-

    ulating thermogenesis, chronic infusion of MCH was shown

    to significantly decrease rectal temperatures and the expres-

    sion of uncoupling protein-1 expression in brown adipose

    tissue [33], whereas mice deficient both in leptin and MCH

    demonstrated an increase in basal body temperature and re-

    sistance to cold exposure [71]. As part of changes elucidated

    by MCH infusion, there is a significant reduction in mRNA

    expression of key molecules involved in fatty acid oxida-

    tion in the brown adipose tissue (BAT), including acyl-CoA

    oxidase and carnitine palmitoyltransferase, suggesting that

    MCH infusion might impair BAT functions [33]. Addition-

    ally, MCH deficiency in ob/ob mice causes a reduction in

    the expression of stearoyl-CoA desaturase-1, another medi-

    ator of the obese phenotype in ob/ob mice [48]. MCH is

    also reported to stimulate leptin mRNA expression and lep-

  • 7/28/2019 Melanina y Emocion

    4/7

    1608 Y. Shi / Peptides 25 (2004) 16051611

    tin secretion [14], and activates signaling pathways in 3T3-

    L1 adipocytes [14,15], suggesting a role in regulating white

    adipocyte function. In addition, MCHR1 expression is also

    reported in insulin-producing RINm5F and CRI-G1 cell-lines

    that are responsive to MCH-stimulated insulin release [75].

    It is not clear whether these peripheral effects are centrally

    mediated by local synthesis of MCH or through paracrineregulation. For example, second-order neurons in both vagal

    and sympathetic innervation of the pancreas are reported to

    contain neuropeptides that control food intake [17].

    4. MCH receptor as a drug target for obesity

    Obesity is a common problem in Western societies and

    is associated with significant morbidity and mortality. Due

    to limited treatment options, the disease represents a large

    unmet medical condition. As a result, pharmaceutical and

    biotech companies have invested significant amount of ef-

    forts and resources in the search for a novel treatment.

    As one of the orexigenic neuropeptides involved in central

    regulation of energy homeostasis, MCH has been subjected

    to intensive scrutinization for its potential as a target for obe-

    sity drug development. A number of highly potent cyclic

    peptide analogs were developed as peptide agonists and an-

    tagonists from structureactivity relationship studies of MCH

    and MCHR [3,68,74]. It was demonstrated that an MCH

    analog consisting of the cyclic core with arginine attached

    to the N-terminus of the disulfide ring was sufficient to acti-

    vate both MCHR1 and MCHR2 as effectively as full-length

    MCH [6] (Fig. 2). The analog was equally effective as MCH

    in inducing food intake in short-term treatment, and adipoc-ity with chronic administration when compared with MCH

    [72,74]. Similarly, a series of analogs with antagonist activ-

    ity were also generated [7]. A potent antagonist was devel-

    oped by replacement of Leu9-Gly10 and Arg14-Pro15 with

    Fig. 2. The structure of MCH and its peptide analogs. Ava, 5-aminovaleric

    acid.

    5-aminovaleric acid, and the analog demonstrated antago-

    nistic activity against MCH [7,72]. When tested in rats, the

    antagonist attenuated the orexigenic effects of an agonist,

    but was ineffective on food intake when given alone [72].

    Development of these peptide analogs provides useful tools

    for exploring physiology mediated by MCHR1, and for fur-

    ther validation of the receptor as a drug target for obesity.However, development of peptide antagonists as a potential

    treatment for obesity faces significant challenges due to their

    required route of delivery acrossthe blood brain barrier, since

    no direct peripheral MCH effects have been reported thus far.

    As a member of the G-protein-coupled receptor super

    family that are amenable to drug screening, cloning of both

    MCHR1 and MCHR2 hasfacilitated the development of cell-

    based assays for high-throughput screening of small MCH

    antagonist molecules. Development of non-peptide antago-

    nists affords direct testing of those compounds for efficacy

    by peripheral injection or feeding without the complica-

    tion associated with ICV injection. Two of the non-peptide

    MCHR1 antagonists discovered by high throughput and tar-geted screenings were recently reported to be effective in

    treating diet-induced obesity in rodents [23] (Fig. 3). The

    first non-peptide antagonist, T-226296, is an orally active

    compound that demonstrated a high affinity and selective

    binding to MCHR1 (Ki = 5.5 nM), but not MCHR2 and

    other homologous receptors [76]. The compound inhibites

    MCH-stimulated arachidonic acid release in heterologous

    expression systems, and also reverses the MCH-mediated in-

    hibition of forskolin-stimulated cAMP accumulation as well

    as MCH-induced Ca2+ release. T-226296 exhibits a good

    oral bioavailability and brain penetrability, and oral admin-

    istration in rats reverses the hyperphagia induced by ICVinjection of exogenous MCH [76]. No further analysis was

    Fig. 3. Chemical structures of two MCHR1 antagonists.

  • 7/28/2019 Melanina y Emocion

    5/7

    Y. Shi / Peptides 25 (2004) 16051611 1609

    reported on the potential of T-226296 on energy expenditure

    and its effectiveness in treating diet-induced obesity.

    A second non-peptide antagonist compound, SNAP-7941,

    was identified by screening of a GPCR-biased compound li-

    brary against MCHR1 in a functional assay measuring in-

    tracellular Ca2+ mobilization [12]. The compound possesses

    more than 1000-fold selectivity for MCHR1 compared withthe human MCHR2 and other related GPCRs. Radio-labeled

    SNAP-7941 specifically binds to MCHR1 in brain sections

    in a pattern that parallels that of the MCHR1 expression in

    differentregions of the brain, including cerebral cortex, olfac-

    tory tubercle, claustrum, and piriform cortex. Chronic injec-

    tion of the compound decreases food intake and body weight,

    andblocks theonset of diet-inducedobesity [12]. The anorec-

    tic effect of SNAP-7941 was unlikely caused by malaise, as

    verified by a taste aversion study. In comparison with fenflu-

    ramine, an antiobesity drug with significant toxic side effects

    [59], the compound causes more sustained weight reduction

    in diet-induced obesity that is much less reversible in weight

    gain after termination of treatment.

    5. MCH and social behaviors

    One of the surprising observations from the treatment of

    rats with SNAP-7941 is the antipsychotic effect of the com-

    pound on mood and emotion, which is supported by binding

    sites of the compound in areas such as the amygdala, accum-

    bens nucleus, dorsal raphe, and locus coeruleus [12,26,62].

    Rats treated with a single oral dose of SNAP-7941 spend

    much less immobile time in the forced-swim test, which

    is comparable with the effect of the typical antidepressantdrug, fluoxitine. The compound is also effective in treating

    anxiety, as evidenced by a significant increase in social

    time of rats treated with 30 mg/kg SNAP-7941 or 5 mg/kg

    chlordiazepoxide, an anxiolytic drug, when compared with

    vehicle-treated controls. These results are consistent with

    hyperactivity observed in MCHR1 knockout mice [20,41],

    but contrast to a previous reports that central administration

    of MCH caused anxiolytic activity [36,42,44]. Further

    analysis is needed to verify whether the antipsychotic

    activity of SNAP-7941 is caused by blocking of the MCH

    activity, or by its off target activity on other unknown brain

    targets. This can be examined by comparing the compound

    with other antagonists of equal efficacy, or by testing the

    compound in MCHR1 knockout mice. In addition, MCH

    has been reported to affect other brain activities, such as

    learning ability and memory retention in rats [42,43,80,81],

    which remain to be validated in the MCHR1 knockout mice

    or with the small molecular antagonists.

    6. Conclusions

    Characterization of MCH in genetically obese rodent

    models and in mice with targeted deletion of MCH and

    MCHR1 firmly establishes a role of the hormone in regulat-

    ing energy homeostasis. MCH, the only neuropeptide in the

    hypothalamus that gives rise to significant changes in energy

    metabolism when ablated in mice, hasattracted significant at-

    tention by the pharmaceutical industry in an effort to develop

    non-peptide antagonists as a treatment for obesity. Demon-

    stration of two small molecular weight antagonists, effectivein treating animal models of obesity, have partially validated

    such an approach, although specificity of these compounds

    remains to be confirmed in mice deficient in MCHR1 expres-

    sion. However,major questions remain aboutthe mechanisms

    regarding the regulatory roles of MCH in energy homeostasis

    and neurological functions. For example, significant differ-

    ences exist between the MCH and MCHR1 knockout mice

    as to the causes for weight loss. There are also differences

    between human and rodents in MCH function as evidenced

    by the lack of a functional MCHR2 receptor in mice and rats.

    Thus, precautions have to be taken when interpreting data

    from rodents in relation to human physiology of MCH me-

    diated events, such as changes in energy expenditure and be-haviors observed in mice with targeted deletion of MCHR1or

    mice treated with antagonist compounds. Furthermore, due

    to a lack of an animal model, it remains a challenge to in-

    vestigate the physiological function of MCHR2 in humans.

    Finally, the wide distribution of MCHR1 expressing neurons

    in the brain suggests multifunctional roles for MCH in regu-

    lating brain activity, such as behavior, olfaction, memory, and

    emotions, which remain to be investigated in future studies.

    Acknowledgements

    The author would like to thank Drs. Jose Caro, Lawrence

    Slieker, and Mark Heiman for insightful comments and help-

    ful suggestions, and John Lockwood for critically reading the

    manuscript.

    References

    [1] Amano M, Takahashi A, Oka Y, Yamanome T, Kawauchi H, Ya-

    mamori K. Immunocytochemical localization and ontogenic devel-

    opment of melanin-concentrating hormone in the brain of a pleu-

    ronectiform fish, the barfin flounder. Cell Tissue Res 2003;311:717.

    [2] An S, Cutler G, Zhao JJ, Huang SG, Tian H, Li W, et al. Iden-

    tification and characterization of a melanin-concentrating hormonereceptor. Proc Natl Acad Sci USA 2001;98:757681.

    [3] Audinot V, Beauverger P, Lahaye C, Suply T, Rodriguez M, Ouvry C,

    et al. Structureactivity relationship studies of melanin-concentrating

    hormone (MCH)-related peptide ligands at SLC-1, the human MCH

    receptor. J Biol Chem 2001;276:1355462.

    [4] Bachner D, Kreienkamp H, Weise C, Buck F, Richter D. Identi-

    fication of melanin concentrating hormone (MCH) as the natural

    ligand for the orphan somatostatin-like receptor 1 (SLC-1). FEBS

    Lett 1999;457:5224.

    [5] Barsh GS, Schwartz MW. Genetic approaches to studying energy

    balance: perception and integration. Nat Rev Genet 2002;3:589600

    (Review).

    [6] Bednarek MA, Feighner SD, Hreniuk DL, Palyha OC, Morin

    NR, Sadowski SJ, et al. Short segment of human melanin-

  • 7/28/2019 Melanina y Emocion

    6/7

    1610 Y. Shi / Peptides 25 (2004) 16051611

    concentrating hormone that is sufficient for full activation of hu-

    man melanin-concentrating hormone receptors 1 and 2. Biochemistry

    2001;40:937986.

    [7] Bednarek MA, Hreniuk DL, Tan C, Palyha OC, MacNeil DJ, Van

    der Ploeg LH, et al. Synthesis and biological evaluation in vitro

    of selective, high affinity peptide antagonists of human melanin-

    concentrating hormone action at human melanin-concentrating hor-

    mone receptor 1. Biochemistry 2002;41:638390.[8] Bednarek MA, Tan C, Hreniuk DL, Palyha OC, MacNeil DJ, Van

    Der Ploeg LH, et al. Synthesis and biological evaluation in vitro

    of a selective, high potency peptide agonist of human melanin-

    concentrating hormone action at human melanin-concentrating hor-

    mone receptor 1. J Biol Chem 2002;277:138216.

    [9] Bittencourt JC, Elias CF. Melanin-concentrating hormone and neu-

    ropeptide E-I projections from the lateral hypothalamic area and zona

    incerta to the medial septal nucleus and spinal cord: a study using

    multiple neuronal tracers. Brain Res 1998;805:119.

    [10] Bittencourt JC, Presse F, Arias C, Peto C, Vaughan J, Nahon JL, et

    al. The melanin-concentrating hormone system of the rat brain: an

    immuno- and hybridisation-histochemical characterization. J Comp

    Neurol 1992;319:21845.

    [11] Bluet-Pajot MT, Presse F, Voko Z, Hoeger C, Mounier F, Epel-

    baum J, et al. Neuropeptide E-I antagonizes the action of melanin-concentrating hormone on stress-induced release of adrenocorti-

    cotropin in the rat. J Neuroendocrinol 1995;7:297303.

    [12] Borowsky B, Durkin MM, Ogozalek K, Marzabadi MR, DeLeon

    J, Lagu B, et al. Antidepressant, anxiolytic and anorectic effects

    of a melanin-concentrating hormone-1 receptor antagonist. Nat Med

    2002;8:82530.

    [13] Borsu L, Presse F, Nahon JL. The AROM gene, spliced mRNAs

    encoding new DNA/RNA-binding proteins are transcribed from the

    opposite strand of the melanin-concentrating hormone gene in mam-

    mals. J Biol Chem 2000;275:4057687.

    [14] Bradley RL, Kokkotou EG, Maratos-Flier E, Cheatham B. Melanin-

    concentrating hormone regulates leptin synthesis and secretion in rat

    adipocytes. Diabetes 2000;49:10737.

    [15] Bradley RL, Mansfield JP, Maratos-Flier E, Cheatham B. Melanin-

    concentrating hormone activates signaling pathways in 3T3-L1adipocytes. Am J Physiol 2002;283:E58492.

    [16] Broberger C, Hokfelt T. Hypothalamic and vagal neuropeptide cir-

    cuitries regulating food intake. Physiol Behav 2001;74:66982.

    [17] Buijs RM, Chun SJ, Niijima A, Romijn HJ, Nagai K. Parasympathe-

    tic and sympathetic control of the pancreas: a role for the suprachi-

    asmatic nucleus and other hypothalamic centers that are involved in

    the regulation of food intake. J Comp Neurol 2001;431:40523.

    [18] Burgaud JL, Poosti R, Fehrentz JA, Martinez J, Nahon JL. Melanin-

    concentrating hormone binding sites in human SVK14 keratinocytes.

    Biochem Biophys Res Commun 1997;241:6229.

    [19] Chambers J, Ames RS, Bergsma D, Muir A, Fitzgerald LR, Hervieu

    G, et al. Melanin-concentrating hormone is the cognate ligand for the

    orphan G-protein-coupled receptor SLC-1. Nature 1999;400:2615.

    [20] Chen Y, Hu C, Hsu CK, Zhang Q, Bi C, Asnicar M, et al. Targeted

    disruption of the melanin-concentrating hormone receptor-1 resultsin hyperphagia and resistance to diet-induced obesity. Endocrinology

    2002;143:246977.

    [21] Courseaux A, Nahon JL. Birth of two chimeric genes in the Ho-

    minidae lineage. Science 2001;291:12937.

    [22] Crowley VE, Yeo GS, ORahilly S. Obesity therapy: altering the

    energy intake-and-expenditure balance sheet. Nat Rev Drug Discov

    2002;1:27686 (Review).

    [23] Forray C. The MCH receptor family: feeding brain disorders? Curr

    Opin Pharm 2003;3:859 (Review).

    [24] Gonzalez MI, Vaziri S, Wilson CA. Behavioral effects of alpha-MSH

    and MCH after central administration in the female rat. Peptides

    1996;17:1717.

    [25] Gottumukkala RV, Waterman EA, Herd LM, Gawkrodger DJ, Watson

    PF, Weetman AP, et al. Autoantibodies in vitiligo patients recognize

    multiple domains of the melanin-concentrating hormone receptor. J

    Invest Dermatol 2003;121:76570.

    [26] Griffond B, Baker BI. Cell and molecular cell biology of

    melanin-concentrating hormone. Int Rev Cytol Surv Cell Biol

    2002;213:23377 (Review).

    [27] Hanada R, Nakazato M, Matsukura S, Murakami N, Yoshimatsu H,

    Sakata T. Differential regulation of melanin-concentrating hormone

    and orexin genes in the agouti-related protein/melanocortin-4 recep-tor system. Biochem Biophys Res Commun 2000;268:8891.

    [28] Harris J, Bird DJ. Modulation of the fish immune system by hor-

    mones. Vet Immunol Immunopathol 2000;77:16376 (Review).

    [29] Hawes BE, Kil E, Green B, ONeill K, Fried S, Graziano MP. The

    melanin-concentrating hormone receptor couples to multiple G pro-

    teins to activate diverse intracellular signaling pathways. Endocrinol-

    ogy 2000;141:452432.

    [30] Herve C, Fellmann D. Changes in rat melanin-concentrating hor-

    mone and dynorphin messenger ribonucleic acids induced by food

    deprivation. Neuropeptides 1997;31:23742.

    [31] Hill J, Duckworth M, Murdock P, Rennie G, Sabido-David C,

    Ames RS, et al. Molecular cloning and functional characteriza-

    tion of MCH2, a novel human MCH receptor. J Biol Chem

    2001;276:201259.

    [32] Hoogduijn MJ, Ancans J, Suzuki I, Estdale S, Thody AJ. Melanin-concentrating hormone and its receptor are expressed and functional

    in human skin. Biochem Biophys Res Commun 2002;296:698 701.

    [33] Ito M, Gomori A, Ishihara A, Oda Z, Mashiko S, Matsushita H, et

    al. Characterization of MCH-mediated obesity in mice. Am J Physiol

    2003;284:E9405.

    [34] Kawano H, Honma S, Honma A, Horie M, Kawano Y, Hayashi S.

    Melanin-concentrating hormone neuron system: the Wide Web that

    controls the feeding. Anat Sci Int 2002;77:14960.

    [35] Kawauchi H, Kawazoe I, Tsubokawa M, Kishida M, Baker BI. Char-

    acterization of melanin-concentrating hormone in chum salmon pi-

    tuitaries. Nature 1983;305:3213.

    [36] Kela J, Salmi P, Rimondini-Giorgini R, Heilig M, Wahlestedt C.

    Behavioural analysis of melanin-concentrating hormone in rats:

    evidence for orexigenic and anxiolytic properties. Regul Pept

    2003;114:10914.[37] Kemp EH, Waterman EA, Hawes BE, ONeill K, Gottumukkala RV,

    Gawkrodger DJ, et al. The melanin-concentrating hormone receptor

    1, a novel target of autoantibody responses in vitiligo. J Clin Invest

    2002;109:92330.

    [38] Lembo PM, Grazzini E, Cao J, Hubatsch DA, Pelletier M, Hof-

    fert C, et al. The receptor for the orexigenic peptide melanin-

    concentrating hormone is a G-protein-coupled receptor. Nat Cell Biol

    1999;1:26771.

    [39] Ludwig DS, Mountjoy KG, Tatro JB, Gillette JA, Frederich RC, Flier

    JS, et al. Melanin-concentrating hormone: a functional melanocortin

    antagonist in the hypothalamus. Am J Physiol 1998;274:E62733.

    [40] Ludwig DS, Tritos NA, Mastaitis JW, Kulkarni R, Kokkotou E,

    Elmquist J, et al. Melanin-concentrating hormone over expression in

    transgenic mice leads to obesity and insulin resistance. J Clin Invest

    2001;107:37986.[41] Marsh DJ, Weingarth DT, Novi DE, Chen HY, Trumbauer ME, Chen

    AS, et al. Melanin-concentrating hormone 1 receptor-deficient mice

    are lean, hyperactive, and hyperphagic and have altered metabolism.

    Proc Natl Acad Sci USA 2002;99:32405.

    [42] Monzon ME, De Barioglio SR. Response to novelty after i.c.v. in-

    jection of melanin-concentrating hormone (MCH) in rats. Physiol

    Behav 1999;67:8137.

    [43] Monzon ME, de Souza MM, Izquierdo LA, Izquierdo I, Barros DM,

    de Barioglio SR. Melanin-concentrating hormone (MCH) modifies

    memory retention in rats. Peptides 1999;20:15179.

    [44] Monzon ME, Varas MM, De Barioglio SR. Anxiogenesis in-

    duced by nitric oxide synthase inhibition and anxiolytic effect

    of melanin-concentrating hormone (MCH) in rat brain. Peptides

    2001;22:10437.

  • 7/28/2019 Melanina y Emocion

    7/7

    Y. Shi / Peptides 25 (2004) 16051611 1611

    [45] Mori M, Harada M, Terao Y, Sugo T, Watanabe T, Shimomura Y, et

    al. Cloning of a novel G protein-coupled receptor, SLT, a subtype of

    the melanin-concentrating hormone receptor. Biochem Biophys Res

    Commun 2001;283:10138.

    [46] Nahon JL. The melanin-concentrating hormone-from the peptide to

    the gene. Crit Rev Neurobiol 1994;8:22162 (Review).

    [47] Nahon JL, Presse F, Bittencourt JC, Sawchenko PE, Vale W. The

    rat melanin-concentrating hormone messenger ribonucleic acid en-codes multiple putative neuropeptides coexpressed in the dorsolateral

    hypothalamus. Endocrinology 1989;125:205665.

    [48] Ntambi JM, Miyazaki M. Recent insights into stearoyl-CoA

    desaturase-1. Curr Opin Lipidol 2003;14:25561 (Review).

    [49] Pandolfi M, Canepa MM, Ravaglia MA, Maggese MC, Paz DA,

    Vissio PG. Melanin-concentrating hormone system in the brain and

    skin of the cichlid fish Cichlasoma dimerus: anatomical localization,

    ontogeny and distribution in comparison to alpha-melanocyte-stimul-

    ating hormone-expressing cells. Cell Tissue Res 2003;311:619.

    [50] Pissios P, Maratos-Flier E. Melanin-concentrating hormone: from fish

    skin to skinny mammals. Trends Endocrinol Metab 2003;14:2438

    (Review).

    [51] Pissios P, Trombly DJ, Tzameli I, Maratos-Flier E. Melanin-

    concentrating hormone receptor 1 activates extracellular signal-

    regulated kinase and synergizes with G(s)-coupled pathways. En-docrinology 2003;144:351423.

    [52] Presse F, Nahon JL, Fischer WH, Vale W. Structure of the

    human melanin concentrating hormone mRNA. Mol Endocrinol

    1990;4:6327.

    [53] Qu D, Ludwig DS, Gammeltoft S, Piper M, Pelleymounter MA,

    Cullen MJ, et al. A role for melanin-concentrating hormone in the

    central regulation of feeding behaviour. Nature 1996;380:2437.

    [54] Rance T, Baker BI. The teleost melanin-concentrating hormone-a

    pituitary hormone of hypothalamic origin. Gen Comp Endocrinol

    1979;37:6473.

    [55] Rees JL. The melanocortin 1 receptor (MC1R): more than just red

    hair. Pigment Cell Res 2000;13:13540 (Review).

    [56] Rodriguez M, Beauverger P, Naime I, Rique H, Ouvry C, Souchaud

    S, et al. Cloning and molecular characterization of the novel hu-

    man melanin-concentrating hormone receptor MCH2. Mol Pharma-col 2001;60:6329.

    [57] Rossi M, Beak SA, Choi SJ, Small CJ, Morgan DG, Ghatei MA,

    et al. Investigation of the feeding effects of melanin concentrat-

    ing hormone on food intake-action independent of galanin and the

    melanocortin receptors. Brain Res 1999;846:16470.

    [58] Rossi M, Choi SJ, OShea D, Miyoshi T, Ghatei MA, Bloom

    SR. Melanin-concentrating hormone acutely stimulates feeding, but

    chronic administration has no effect on body weight. Endocrinology

    1997;138:3515.

    [59] Rothman RB, Baumann MH. Therapeutic adverse actions of seroto-

    nin transporter substrates. Pharmacol Ther 2002;95:7388 (Review).

    [60] Sahu A. Leptin decreases food intake induced by melanin-

    concentrating hormone (MCH), galanin (GAL) and neuropeptide Y

    (NPY) in the rat. Endocrinology 1998;139:473942.

    [61] Sailer AW, Sano H, Zeng Z, McDonald TP, Pan J, Pong SS,et al. Identification and characterization of a second melanin-

    concentrating hormone receptor, MCH-2R. Proc Natl Acad Sci USA

    2001;98:75649.

    [62] Saito Y, Cheng M, Leslie FM, Civelli O. Expression of the melanin-

    concentrating hormone (MCH) receptor mRNA in the rat brain. J

    Comp Neurol 2001;435:2640.

    [63] Saito Y, Nothacker HP, Civelli O. Melanin-concentrating hormone

    receptor: an orphan receptor fits the key. Trends Endocrinol Metab

    2000;11:299303 (Review).

    [64] Saito Y, Nothacker HP, Wang Z, Lin SH, Leslie F, Civelli O. Molec-

    ular characterization of the melanin-concentrating hormone-receptor.

    Nature 1999;400:2659.

    [65] Saito Y, Wang Z, Hagino-Yamagishi K, Civelli O, Kawashima S,

    Maruyama K. Endogenous melanin-concentrating hormone receptor

    SLC-1 in human melanoma SK-MEL-37 cells. Biochem Biophys

    Res Commun 2001;289:4450.

    [66] Saper CB, Chou TC, Elmquist JK. The need to feed: homeostatic

    and hedonic control of eating. Neuron 2002;36:199211.

    [67] Schaffer JV, Bolognia JL. The melanocortin-1 receptor-red hair and

    beyond. Arch Dermatol 2001;137:147785 (Review).

    [68] Schlumberger SE, Talke-Messerer C, Zumsteg U, Eberle AN. Ex-

    pression of receptors for melanin-concentrating hormone (MCH)in different tissues and cell lines. J Recept Signal Transduct Res

    2002;22:50931.

    [69] Schwartz MW, Gelling RW. Rats lighten up with MCH antagonist.

    Nat Med 2002;8:77981.

    [70] Schwartz MW, Woods SC, Porte D, Seeley RJ, Baskin DG. Cen-

    tral nervous system control of food intake. Nature 2000;404:66171

    (Review).

    [71] Segal-Lieberman G, Bradley RL, Kokkotou E, Carlson M, Trombly

    DJ, Wang X, et al. Melanin-concentrating hormone is a critical me-

    diator of the leptin-deficient phenotype. Proc Natl Acad Sci USA

    2003;100:1008590.

    [72] Shearman LP, Camacho RE, Stribling DS, Zhou D, Bednarek

    MA, Hreniuk DL, et al. Chronic MCH-1 receptor modulation al-

    ters appetite, body weight and adiposity in rats. Eur J Pharmacol

    2003;475:3747.[73] Shimada M, Tritos NA, Lowell BB, Flier JS, Maratos-Flier E. Mice

    lacking melanin-concentrating hormone are hypophagic and lean.

    Nature 1998;396:6704.

    [74] Suply T, Della Zuana O, Audinot V, Rodriguez M, Beau-

    verger P, Duhault J, et al. SLC-1 receptor mediates effect of

    melanin-concentrating hormone on feeding behavior in rat: a

    structureactivity study. J Pharmacol Exp Ther 2001;299:13746.

    [75] Tadayyon M, Welters HJ, Haynes AC, Cluderay JE, Hervieu

    G. Expression of melanin-concentrating hormone receptors in

    insulin-producing cells: MCH stimulates insulin release in

    RINm5F and CRI-G1 cell-lines. Biochem Biophys Res Commun

    2000;275:70912.

    [76] Takekawa S, Asami A, Ishihara Y, Terauchi J, Kato K, Shimo-

    mura Y, et al. T-226296: a novel, orally active and selective

    melanin-concentrating hormone receptor antagonist. Eur J Pharmacol2002;438:12935.

    [77] Tan CP, Sano H, Iwaasa H, Pan J, Sailer AW, Hreniuk DL, et al.

    Melanin-concentrating hormone receptor subtypes 1 and 2: species-

    specific gene expression. Genomics 2002;79:78592.

    [78] Toumaniantz G, Bittencourt JC, Nahon JL. The rat melanin-

    concentrating hormone gene encodes an additional putative protein

    in a different reading frame. Endocrinology 1996;137:451821.

    [79] Tritos NA, Vicent D, Gillette J, Ludwig DS, Flier ES, Maratos-Flier

    E. Functional interactions between melanin-concentrating hormone,

    neuropeptide Y, and anorectic neuropeptides in the rat hypothalamus.

    Diabetes 1998;47:168792.

    [80] Varas M, Perez M, Monzon ME, de Barioglio SR. Melanin-

    concentrating hormone, hippocampal nitric oxide levels and memory

    retention. Peptides 2002;23:221321.

    [81] Varas M, Perez M, Ramirez O, de Barioglio SR. Melanin concen-trating hormone increase hippocampal synaptic transmission in the

    rat. Peptides 2002;23:1515.

    [82] Vaughan JM, Fischer WH, Hoeger C, Rivier J, Vale W. Character-

    ization of melanin-concentrating hormone from rat hypothalamus.

    Endocrinology 1989;125:16605.

    [83] Voisey J, Van Daal A. Agouti: from mouse to man, from skin to fat.

    Pigment Cell Res 2002;15:108 (Review).

    [84] Wang S, Behan J, ONeill K, Weig B, Fried S, Laz T, et al. Iden-

    tification and pharmacological characterization of a novel human

    melanin-concentrating hormone receptor, MCH-R2. J Biol Chem

    2001;276:3466470.

    [85] Yu HS. Melanocyte destruction and repigmentation in vitiligo:

    a model for nerve cell damage and regrowth. J Biol Chem

    2002;9:56473 (Review).