activation of the hypothalamic

Upload: bhartinder

Post on 06-Mar-2016

6 views

Category:

Documents


0 download

DESCRIPTION

Addictive drugs (opiates, ethanol, cannabinoids (CBs),nicotine, cocaine, amphetamines) induce activation ofthe hypothalamic–pituitary–adrenal (HPA) axis, with thesubsequent release of adrenocorticotropic hormone andglucocorticoids. The sequence of events leading to HPAactivation appears to start within the brain, suggestingthat activation is not secondary to peripheral homeostaticalterations. The precise neurochemical mechanismsand brain pathways involved are markedlydependent on the particular drug, although it is assumedthat information eventually converges into the hypothalamicparaventricular nucleus (PVN). Whereas somedrugs may act on the hypothalamus or directly withinPVN neurons (i.e. ethanol), others exert their primaryaction outside the PVN (i.e. CBs, nicotine, cocaine).Corticotropin-releasing hormone (CRH) has a critical rolein most cases, but the changes in c-fos and CRH geneexpression in the PVN also reveal differences amongdrugs. More studies are needed to understand howaddictive drugs act on this important neuroendocrinesystem and their functional consequences.IntroductionThe hypothalamic–pituitary–adrenal (HPA) axis is a

TRANSCRIPT

  • Activation of the hypothalamicpituitaryadrenal axis by addictivedrugs: different pathways, commonoutcomeAntonio Armario

    Institute of Neurosciences and Animal Physiology Unit (Department of Cellular Biology, Physiology and Immunology),Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain

    Addictive drugs (opiates, ethanol, cannabinoids (CBs),nicotine, cocaine, amphetamines) induce activation ofthe hypothalamicpituitaryadrenal (HPA) axis, with thesubsequent release of adrenocorticotropic hormone andglucocorticoids. The sequence of events leading to HPAactivation appears to start within the brain, suggestingthat activation is not secondary to peripheral homeo-static alterations. The precise neurochemical mechan-isms and brain pathways involved are markedlydependent on the particular drug, although it is assumedthat information eventually converges into the hypo-thalamic paraventricular nucleus (PVN). Whereas somedrugs may act on the hypothalamus or directly withinPVN neurons (i.e. ethanol), others exert their primaryaction outside the PVN (i.e. CBs, nicotine, cocaine).Corticotropin-releasing hormone (CRH) has a critical rolein most cases, but the changes in c-fos and CRH geneexpression in the PVN also reveal differences amongdrugs. More studies are needed to understand howaddictive drugs act on this important neuroendocrinesystem and their functional consequences.

    IntroductionThe hypothalamicpituitaryadrenal (HPA) axis is a com-plex neuroendocrine system involved in an importantnumber of central and peripheral physiological functions,most of them related to appropriate adaptation to stressfulsituations. The HPA axis is also altered in an importantnumber of pathophysiological and psychopathological pro-cesses, including drug addiction [1,2]. The main features ofthe organization of the HPA axis and brain areas control-ling it are well-known [3,4]. In brief, theHPA axis ismainlygoverned by the paraventricular nucleus (PVN) of thehypothalamus, particularly the medial dorsal parvocellu-lar region (mpdPVN). In this region, there is rich popu-lation of parvocellular neurons expressing a wide range ofneuropeptides. The most important neuropeptides for theregulation of the HPA axis are the corticotrophin-releasingfactor or hormone (CRF or CRH) and vasopressin (arginin-vasopressin (AVP) in most mammals, including rats andhumans). AVP is formed in a percentage of CRHergicneurons and the number of CRH+ neurons also expressing

    Review

    Glossary of terms

    ACTH: adrenocorticotropic hormone, the hormone synthesized in the anterior

    pituitary gland that controls glucocorticoid secretion in the adrenal gland.

    A1/C1: refers to a cluster of neurons using noradrenaline (A1) or adrenaline

    (C1) as neurotransmitters, which are within the anatomically defined

    ventrollateral medulla.

    A2/C2: refers to a cluster of neurons using noradrenaline (A2) or adrenaline

    (C2) as neurotransmitters, which are within the anatomically defined nucleus

    tractus solitarius.

    AP: anterior pituitary (hypophysis), part of the hypophysis where an important

    number of hormones (including ACTH) are formed.

    AVP: arginin-vasopressin, a neuropeptide formed in the supraoptic and

    paraventricular nuclei of the hypothalamus that participates in the control of

    ACTH in the anterior pituitary.

    BBB: bloodbrain barrier, the barrier that limits entry into the brain of

    peripheral hydrophilic molecules.

    BLA: basolateral amygdala

    CB: cannabinoids, the exogenous and endogenous (eCBs) substances that

    interact with cannabinoid receptors.

    CB1, CB2: the two well-characterized types of receptors for cannabinoids.

    CRH (or CRF): corticotropin-releasing hormone, the most important hypotha-

    lamic stimulatory factors for the control of ACTH.

    DARPP-32: dopamine- and cAMP-dependent phosphoprotein of 32 kDa; it is a

    protein activated by dopamine and other neurotransmitters which inhibits

    phosphatases.

    DOR: d (delta) opioid receptors

    eEOPs: endogenous opioids, the endogenous peptides that interact with opioid

    receptors.

    GR: glucocorticoid (type II) receptors, a type of genomic receptors for

    glucocorticoid hormones.

    HPA: hypothalamicpituitaryadrenal axis; refers to the neuroendocrine

    system that controls glucocorticoid secretion by the adrenal gland.

    IEGs: immediate early genes, genes that are expressed immediately in

    response to stimuli and are markers of neuronal activation.

    KOR: k (kappa) opioid receptors

    MDMA: 3,4-methylenedioximetamphetamine.

    ME: median eminence, part of the hypophysis where the capillaries of the

    pituitary portal blood are located.

    MOR: m (mu) opioid receptors

    MR: mineralocorticoid (type I) receptors, a type of genomic receptor for

    glucocorticoid hormones.

    NGFI-B: a transcription factor rapidly induced by the neurotrophin nerve

    growth factor, which is considered to be an immediate early gene.

    NMDA: n-methyl-D-aspartate, defines a subtype of the ionotropic glutamate

    receptor.

    PDYN: pro-dynorphin; refers to the gene and precursor molecule of some

    endogenous opioids, mainly dynorphin.

    PENK: pro-enkephalin; refers to the gene and the precursor molecule of the

    endogenous opioids enkephalins.

    POMC: pro-opiomelanocortin; refers to the gene and the precursor molecule of

    ACTH and b-endorphin.

    PPB: pituitary-portal blood, a special circulatory system that collects sub-

    stances released by hypothalamic axons and directs them to the anterior

    pituitary.

    PVN: paraventricular nucleus of the hypothalamus, the key nucleus in the

    control of the HPA axis. The PVN has several subregions, including the medial

    parvocellular dorsal (mpdPVN), whose neurons send axons to the externalCorresponding author: Armario, A. ([email protected]).318 0165-6147/$ see front matter 2010 Elsevier Ltd. All rights reserved. doi:10.1016/j.tips.2010.04.005 Trends in Pharmacological Sciences 31 (2010) 318325

  • for CRH (and sometimes for AVP) can be detected in thempdPVN 24 h after initial exposure to stress. However,more sensitive tools such as intronic probes (which detectnuclear primary transcripts (hnRNA)), are needed to ana-lyze the situation after brief exposures or low intensitystressors [5,6]. Exposure to stress can result in increasedgene expression of pro-opiomelanocortin (POMC), the pre-cursor of ACTH that also gives rise to endogenous opioids(eOPs) such as b-endorphin in the AP.

    In addition to the different time-course of ACTH andcorticosterone secretion, evaluation of the effects of stress(or drugs) based only on glucocorticoids can lead to mis-leading interpretations. Adrenocortical secretion saturateswith intermediate levels of plasma ACTH, so plasma levelsof corticosterone may not reflect actual ACTH release ifonly a single time-point is evaluated [4]. In addition, thereis evidence for an actual dissociation after exposure toprolonged stress (usually >6 h) when almost normal levelsof ACTH can be observed with high levels of corticosterone[7]. We do not have a direct explanation for this phenom-enon, but we know that sympathetic neural innervation ofthe adrenal gland can modulate adrenal sensitivity tocirculating ACTH [8] and this may explain (at least inpart) some cases of partial dissociation between the twohormones. Finally, locally generated signals as well asother circulating factors can contribute, under certainconditions, to the release of glucocorticoids, although inall cases the presence of a minimum amount of ACTHappears to be critical. Glucocorticoids released duringstress have a wide impact on peripheral tissues and the

    type-II or glucocorticoid receptors (GR). In addition, thereis evidence for fast non-genomic effects of glucocorticoidthrough putative transmembrane receptors. One criticalaction of glucocorticoids is the negative feedback they exertat multiple levels within the brain and the AP.

    Within the context of stress theories, it is unclear why somany drugs can activate the HPA axis [4]. Although theeffects of some drugs may be secondary to homeostaticalterations, it is more likely that they are related to thepharmacological activation of brain pathways involved inthe response to stressors. A general scheme of the placewhere addictive drugs and other stimuli can initiate theevents leading to the activation of the HPA axis can be seenin Figure 1. Controversial results are more frequentlyencountered in in vitro studies that evaluate the directeffects of addictive drugs on the hypothalamus, AP oradrenal cortex than in in vivo studies. This is probablydue to important problems inherent to in vitro prep-arations.

    Due to space limitations, the present review will focuson how acute exposure to addictive drugs activates theHPA axis. I shall exclude the interesting topics of long-term exposure to drugs in early life and the consequences ofchronic exposure to drugs (including tolerance and with-drawal) in adults. There is evidence that all addictivedrugs can activate the HPA axis in humans with theexception of opiates, which have a predominant inhibitoryrole in humans [11]. Putative mechanisms have beenstudied in laboratory animals, so specific references tohuman studies will not bemade. Tables 1 and 2 summarizeAVP is increased after chronic activation of the HPA axis[4]. AVP, acting through V1b receptors, exerts a weakstimulation of corticotropes but markedly potentiatesthe effects of CRH. In addition to CRH and AVP, otherhypothalamic factors can regulate the HPA axis. Axons ofparvocellular mpdPVN neurons terminate around thecapillaries of the pituitary portal blood (PPB) of themedianeminence (ME) where they release CRH, AVP and otherneuropeptides. These reach the anterior pituitary (AP) toactivate adrenocorticotropic hormone (ACTH) release bycorticotrope cells. CRH acts in the AP through type-1receptors (CRH-R1). ACTH then activates the synthesisand secretion of glucocorticoids (cortisol in humans andmost mammals; corticosterone in rodents) in the zonafasciculata of the adrenal cortex throughmelanocortin type2 receptors. Maximum release of ACTH is usually observed510 min after the start of exposure to stressors, whereasmaximum corticosterone levels are achieved after only 2030 min.

    Stimuli that activate the release of HPA hormones notonly induce the release of CRH, AVP and other putativesecretagogues into the PPB, but also increase transcriptionof those genes in the mpdPVN. When the stressors are ofsufficient magnitude and exposure to them is relativelylong (30 min to several hours), increased levels of mRNA

    median eminence, and the magnocellular (mPVN), whose neurons send axonsto the neurohypophysis.

    SA: self-administration; refers to the procedure that allow animals to self-

    administer the drug using operant learning.

    THC: delta9-tetrahydrocannabinol, the major psychotropic component of

    cannabis.

    Reviewbrain [9,10], acting through two types of genomic receptors:type-I or mineralocorticoid receptors (MR) receptors and

    Figure 1. Putative pathways whereby drugs can activate different components of

    the HPA axis. Drugs can act at the following sites: 1) peripherally, by inducing

    homeostatic alterations or other peripheral changes (i.e. release of other

    substances) that can then activate receptors located in neurons of the

    autonomous nervous system. These neurons in turn could inform the brain

    through synapses in the nucleus tractus solitarius (NTS) or adjacent regions; 23)

    on brain areas (NTS, VLM) that send direct inputs to the PVN; 4) on

    circumventricular areas that are outside the BBB (i.e, organum vasculosum

    lamina terminals or subfornical organ (SFO); 5) on receptors located in specific

    brain regions, proximal (i.e. other hypothalamic nuclei) or more distal (i.e. limbic

    areas) to the PVN, thus activating circuits that finally would convey into the PVN; 6)

    directly on PVN neurons or even on CRH neurons; 7) on the ME, directly controlling

    the release of hypothalamic factors from axon terminals; 8) on the AP, modulating

    the response of corticotropes to hypothalamic regulatory factors; 9) on the zona

    fasciculata of the adrenal cortex or on the brain areas controlling the sympathetic

    innervation of the adrenal, in both cases modulating the response to ACTH.

    Trends in Pharmacological Sciences Vol.31 No.7319

  • f ad

    c

    C

    Y

    Y

    Y

    sive

    nvo

    dial

    erethe main results that I elaborate on and which may assistthe reader in following the discussion. Among the drugs tobe reviewed, some of them are classically considered asdepressants (opiates, ethanol, CBs), whereas the othersare psychostimulants (nicotine, cocaine, amphetamines

    Table 1. Summary of the knowledge about the acute effects o

    Drug Inside

    BBB

    PVN

    lesions

    IEGs

    PVN

    Opiates Yesa Yes

    Ethanol Yes Yes Yes

    Cannabinoids Yes Yes

    Nicotine Yes Yes

    Cocaine Yes Yes No

    Amphetamine Yes

    a: Yes indicates positive evidence and No negative evidence; ? indicates no conclu

    Table 2. Brain areas and neurotransmitters/neuromodulators i

    Drug Primary site(s) of action

    Opiates Hypothalamus

    Ethanol HypothalamusPVN?

    Cannabinoids Basolateral amygdala, Me

    Nicotine Nucleus tractus solitarius

    Cocaine Ventral striatum

    Amphetamine and derivatives

    ? indicates a possible, but not conclusive, site of action; empty squares indicate that th

    Reviewand their derivatives).

    Opiates and eOPsThere are three families of eOPs, each one coded by a singlegene: proopiomelanocortin (POMC), proenkephalin(PENK) and prodynorphin (PDYN). Each protein precur-sor can give rise to different peptides, somewith affinity fordifferent opioid receptors. b-endorphin (derived fromPOMC) has preferential affinity for m receptors (MOR),leu and met-enkephalin (derived mainly from PENK) havepreferential affinity for d receptors (DOR) and dynorphins(derived from PDYN) has preferential affinity for kreceptors (KOR). Opiate drugs (i.e. morphine, heroin,methadone) are predominantly agonists of MOR, whereasnaloxone is a well-characterized opioid receptor antagonistwith affinity for all types of receptors, but particularly forMOR. Naltrexone has similar properties as naloxone, but alonger-lasting action. Most of the data on the role of opiatesand eOPs on neuroendocrine regulation were obtained inthe 1960s and 1980s, and the interest subsequentlydeclined, although very recently some of these effects havebeen reviewed [11]. Whereas in rodents eOPs may exertstimulatory and inhibitory roles in the control of the HPAaxis, they have a predominantly inhibitory role in humans,with consequent activation of the HPA axis after admin-istration of naloxone/naltrexone [11]. Exposure to opiatescan therefore have clearly different consequences on theHPA axis and physiological functions affected by the HPAaxis in humans as compared with rodents.

    320Early reports demonstrated that morphine can activateadrenocortical secretion through a mechanism dependentupon the hypothalamus and the AP [12]. Although somedirect effects at the adrenal level have been reported [12],the main effects are exerted above the AP. Methadone also

    dictive drugs on the HPA axis

    -fos in PVN

    RF neurons

    PVN CRF gene

    expression

    Role

    of CRF

    Role

    of AVP

    es ? Yes

    Yes Yes Yes

    Yes

    es

    Yes Yes No

    es Yes

    evidence; empty squares indicate that no studies have been done.

    lved in the effects of addictive drugs on the HPA axis

    Neurotransmitters/neuromodulators involved

    amygdala Noradrenaline, Serotonin

    Noradrenaline/adrenalineGlutamate (NMDA)Nitric oxideCRHDopamineNoradrenaline/adrenalineAcetylcholine (muscarinic)Glutamate (NMDA)Endogenous opioidsCRHSerotonin

    are no studies or positive evidence for any site or neurotransmitter/neuromodulator.

    Trends in Pharmacological Sciences Vol.31 No.7activates the HPA axis in rats dose-dependently, the effectbeing blocked by naloxone [13]. In rats, activation of theHPA axis was observed with agonists of the three types ofopioid receptors when given peripherally and centrally[1416]. The critical role of MOR in the effects of morphineis supported by the lack of effect of the drug on corticos-terone levels in MOR knockout mice [17]. An independenteffect of each type of receptor on HPA activation is sup-ported by the lack of cross-tolerance after repeated admin-istration [16].

    A direct hypothalamic effect of opioids is supported bythe fact that morphine and enkephalins can increase invitro hypothalamic CRH secretion, an effect blocked bynaloxone [18]. Prior administration of an antiserumagainst rat CRH completely blocked HPA activationcaused by the KOR agonist MR-2034, whereas it onlypartially reduced the response to morphine [19]. Thespecific contribution of CRH to the activation induced byDOR is not known. Despite the involvement of CRH,neither acute nor chronic peripheral morphine adminis-tration altered CRH mRNA levels in the mpdPVN,although enhanced expression was found after naloxone-precipitated withdrawal [20]. Consistent with these find-ings, six repeated administrations of morphine every twohours did not increase CRH gene expression in the PVN, orPOMC gene expression in the AP [21]. There is evidencethat agonists of MOR and KOR (morphine and U-50,488H,respectively) induce c-fos expression in the PVN [22], soopiates can promote expression of the immediate early

  • gene (IEG) c-fos without affecting expression of the CRHgene. However, results with intronic probes are needed toconfirm the lack of effect of opiates in CRHgene expression.Interestingly, intracerebroventricular (i.c.v.) adminis-tration of b-endorphin in rats not only increased plasmaACTH, but also c-fos and CRH gene expression in the PVN[23]. This could be because the effect of b-endorphin wassufficient to be detected with CRHmRNA levels or becausethe peptide and some of its in vivo-generated fragmentsalso activated DOR [24].

    Although weak MOR receptor expression and strongerKOR expression is observed in the PVN [25], there is nodirect evidence for the presence of some types of opioidreceptor specifically in CRH neurons. Similarly, in vivostudies on the local PVN action of opioids have not beenreported. It is therefore possible that the primary effects ofopioids are exerted outside the PVN and perhaps in part atthe level of the ME, where the presence of MOR has beenclearly identified [25].

    Naloxone at relatively high doses activates the HPAaxis [12,26], suggesting a dual role of eOPs in regulation oftheHPA axis. The effects of naloxone are intriguing in that:(i) naloxone methylbromide, an analog that cannot crossthe bloodbrain barrier (BBB), can enhance ACTHsecretion [27]; (ii) its effect was maintained after totalhypothalamic deafferentation and blocked by the syntheticglucocorticoid dexamethasone [28]; and (iii) it was blockedby prior CRH antiserum [19], although the drug did notstimulate in vitro CRH release [18]. Considering thepossible mechanisms outlined in Figure 1, the drug mayact directly on hypothalamic nuclei located outside theBBB that have direct or indirect connections to the PVN.

    EthanolPeripheral ethanol administration activates the HPA axisin laboratory animals and humans. Most of our knowledgeabout how it acts is based on the great contribution of theRivier research team, who reviewed the topic in 1996 [29].Ethanol self-administration (SA) modestly increasesplasma ACTH [30]. Although some effect at the level ofthe AP or the adrenal gland has been described [29], amajor contribution of these actions to plasma levels of HPAhormones is unlikely. In fact, recent evidence with i.c.v.administration demonstrates that ethanol mainly acts inthe brain to activate theHPA axis [31]. Interestingly, whileinducing similar levels of ACTH, the intraperitoneal (i.p.)administration preferentially activates mpdPVN c-fos andCRH gene expression as compared to oral (p.o.) adminis-tration [32]. Whereas there is evidence that some of thebehavioral effects of ethanol are due to the generation ofacetaldehyde from ethanol by catalase rather than toethanol [33], the results regarding the HPA axis are con-troversial [34,35].

    An important number of studies have delineated themain effects of ethanol, demonstrating [29]: (i) a reductionof the response after PVN lesions; (ii) an enhanced expres-sion of IEGs c-fos and NGFI-B; (iii) an increase in PVNCRH gene expression, which was observed with intronicprobes; and (iii) amodest contribution of AVP. In support of

    Reviewa contribution of both peptides, the HPA response toethanol is completely blocked by i.v. administration of aCRH antiserum [36], basically disappears in mice null forthe CRH-R1 [37], and is reduced in V1b knockoutmice [38].An AVP antiserum reduced the ACTH response to ethanolnot only in control, but also in PVN-lesioned rats [39], so acontribution of AVP neurons outside the PVN is likely.Unfortunately, the origin of AVP neurons that participatein ethanol-induced activation of the HPA axis is unclear. Inany case, CRH appears to be the main mediator of ethanol,which acts on the hypothalamus, perhaps directly on CRHneurons [40]. The i.c.v. administration increased the PVNCRH primary transcript, in accordance with previous datausing peripheral administration. In addition, i.p. and i.c.v.administration increased plasma ACTH and AP POMCgene expression, and these effects were completely blockedby simultaneous administration of CRH and AVP antisera[31].

    CBsD9-tetrahydrocannabinol (THC) is the major psychotropiccomponent ofCannabis sativa and themost studied CB. CBdrugs interact with CB1 and CB2 receptors, the formerbeing the most expressed CB receptor in the brain.Endogenous cannabinoids (eCBs) such as anandamide(N-arachidonoyl-ethanolamine) and 2-arachidonoyl-gly-cerol have affinity for CB1 and CB2 receptors. From the1970s, evidence has accumulated about a stimulatory effectof THC and other CB1 agonists on the HPA axis in severalspecies [41]. CBs exert their effects on the HPA axis actingmainly in the brain as demonstrated by i.c.v. adminis-tration of THC and anandamide [42,43]. The effect ofTHC is not observed in hypothalamic-deafferented rats[44], so the primary effect should be outside the hypothala-mus, although the PVN and the CRHappear to be involved.

    Exogenous CBs and eCBs increase c-fos expression andCRH mRNA levels in the PVN [41]. Nevertheless, directevidence for a role of CRH (i.e. using peripheral adminis-tration of CRH antisera or CRH antagonists) is lacking.The finding that THC-induced release of HPA hormones islikely to be initiated in extra-hypothalamic regions raisessome questions about the role of CB receptors in the PVN.Double in-situ procedures have shown that CB1 receptorsare expressed in >50% of CRH neurons of the PVN [45]. Ifsuch CB1 receptors were demonstrated to be localized inthe terminals of CRH neurons, they could exert an inhibi-tory presynaptic role on ME CRH release after exposure tostress or other stimuli (see below), but a stimulatory role atthis level is unlikely.

    Consistent with a stimulatory role of CBs on HPAactivity, administration of AM404 (eCB reuptake inhibi-tor) and URB597 (inhibitor of the eCB degrading enzymefatty acid amide hydrolase), increased plasma corticoster-one in mice [46] and the effects were blocked by the CB1antagonist AM251. This is important because under cer-tain conditions, eCBs would not activate the HPA axisthrough CB1. Thus, administration of anandamide wasfound to induce release of HPA hormones and c-fos expres-sion in the PVN; neither of the two effects was blocked bythe CB1 antagonist SR 141716 [47]. In addition, activationof the HPA by anandamide is observed in CB1/ mice

    Trends in Pharmacological Sciences Vol.31 No.7[48]. The precise receptors involved in the effects of exogen-ously administered anandamide on the HPA axis are not

    321

  • known, but it is important to be certain that brain levels ofanandamide are within the physiological range to demon-strate a putative biological significance.

    Another intriguing aspect of CBs and the HPA axis isthat CB1 antagonists can also activate the HPA axis dose-dependently. This has been reported with SR141716 afterperipheral and i.c.v. administration in rats and mice[43,4951]. These data suggest a tonic inhibitory role ofeCBs on HPA activity that is compatible with the fact thatSR141716 potentiated corticosterone response to restraintand restraint-induced c-fos expression in the PVN,whereas the opposite was found after potentiation of eCBs[50]. The stimulatory effect of CB1 antagonists on the HPAaxis was not observed in CB1/ mice [50,51], supportingthe hypothesis of mediation by CB1 receptors.

    The data detailed above strongly indicate that CB1exerts a dual effect on the regulation of the HPA axis.CB1/ mice showed higher CRH mRNA levels in thePVN than wild-type mice [45], so it is possible that theinhibitory effects of CB1 receptors predominate overstimulatory effects or that inhibition has a tonic componentand stimulation a phasic one. The inhibitory effects of CB1on HPA activation are compatible with the electrophysio-logical evidence that eCBs may participate in fast negativeglucocorticoid feedback in the PVN through presynapticprotein G-dependent modulation of glutamate release onexcitatory synapses on CRH neurons [52]. However,additional evidence for a functional role of eCBs on nega-tive glucocorticoid feedback is lacking.

    Recent studies shed light on the extra-PVN locationswhere eCBs could act to initiate activation of the HPA axis.Local administration of both the CB1 agonistWIN55,212-2and the CB1 antagonist AM251 in the basolateral amyg-dala (BLA) increased plasma corticosterone in rats [53].These results indicate that the BLA may be one of thestructures where a double function of CB1 on HPA regu-lation is operating. The authors also observed thatexposure to stress after WIN did not further increasecorticosterone levels above those observed with WIN andthey interpret these data as evidence for a role of CB1receptors to inhibit stress-induced HPA activation. This issupported by another report in which activation of CB1 inthe BLA with local administration of a dose of the agonistHU-210 that per se had no effect markedly reduced thecorticosterone response to restraint stress [54]. No effectwas observed in the central amygdala whereas, in themedial amygdala, HU210 enhanced the response torestraint. The different results obtained with HU210 ver-sus WIN injected within the BLA under non-stress con-ditions cannot be explained, although interaction of thesedrugs with receptors other than CB1 may be considered.Nevertheless, we may tentatively conclude that eCBs mayhave a dual role within the BLA to control the restingactivity of theHPA axis, whereas eCBswithin the BLA andthe medial amygdala may have important (but opposite)roles in stress-induced HPA activation. All these data arecompatible with an important extra-PVN role of eCBs tocontrol the HPA axis. Corticosterone response to the CB1agonist HU-210 was not blocked by i.c.v. administration of

    Reviewthe CRH antagonist d-Phe CRH1241 [55], suggesting noextra-pituitary actions of CRH. Recent evidence suggests a

    322partial role of noradrenergic and serotoninergic trans-mission [56].

    NicotineAfter early reports of increases in plasma glucocorticoidsafter nicotine administration in dogs and rats, Cam et al.[57] demonstrated a dose-dependent response that wascompletely abolished in hypophysectomized rats, makingan ACTH-independent effect at the level of the adrenalcortex unlikely. Nicotine levels achieved during SA pro-cedures are sufficient to modestly increase HPA hormones[58,59]. The central action of nicotine and the critical role ofthe PVN have been demonstrated with i.c.v. adminis-tration of nicotine receptor antibodies and bilateral PVNlesions that blocked the HPA response [60]. Low doses ofnicotine preferentially induce c-fos expression in thempdPVN, whereas higher doses also activate the magno-cellular region (mPVN) [61], andmost of mpdPVN neuronsactivated are CRH+ [62,63]. However, there is no report onthe acute effects of the drug on PVN CRH gene expression,and no direct evidence for a critical role of CRH at the AP.

    Sharp and colleagues made the most important contri-butions to the characterization of the brain mechanismsinvolved in nicotine-induced activation of the HPA axis[64]. They conclusively confirmed (using agonists andantagonists that can or cannot cross the BBB) that themain action of nicotine is within the brain, with noadditional effect at the level of the AP [65]. The drugwas less effective when given around the PVN than inthe fourth ventricle [66], ruling out a major direct actionwithin the PVN. The PVN is innervated by noradrenergicand adrenergic neurons from the nucleus tractus solitarius(corresponding to A2/C2 areas) and the ventrollateralmedulla (corresponding to A1/C1) and, to a lesser extent,by locus coeruleus (LC). The authors demonstrated that alow peripheral dose of nicotine activated (in terms of c-fos)the mpdPVN and the A2/C2 region, whereas higher dosesadditionally activated A1/C1, LC and mPVN [61]. Accord-ingly, after local injection of nicotine and the nicotinicantagonist mecamylamine, they concluded that the mainlocus of action was the A2/C2 region [67]. Although only aminority of neurons activated in these regions was tyro-sine-hydroxylase+ [68], there is evidence that nicotine-induced release of noradrenaline in the PVN, actingthrough a1 and a2 but not b-adrenoceptors, is involvedin HPA activation [69,70]. Nicotine-induced activation ofA2/C2 neurons appears to be exerted through bungaro-toxin-insensitive nicotinic receptors [71] and requires thelocal action of glutamate through NMDA receptors andnitric oxide [72]. Administration via i.c.v. means of a CRH-R1 antagonist (CP-154,526) has demonstrated a role ofcentral CRH through CRH-R1 in nicotine-induced releaseof noradrenaline in the PVN and HPA activation [73],although the precise sites of action remain unknown.

    Cocaine and amphetamine-like drugsThe main action of cocaine is to inhibit the reuptake ofmonoamines (dopamine, noradrenaline, serotonin). Acuteadministration of cocaine has been repeatedly found to

    Trends in Pharmacological Sciences Vol.31 No.7dose-dependently activate the HPA axis [74,75]. Achievinga supra-threshold level of cocaine rather than the total

  • amount of drug entering the circulation in a period of timeappears to be critical because cocaine administration withosmotic minipumps did not result in HPA activation [76].Cocaine levels achieved after drug SA also increased corti-costerone [77,78], although whether the increase is higherafter contingent than non-contingent administration iscontroversial [79,80]. A central action of cocaine is sup-ported by the increase in HPA hormones observed afteri.c.v. administration [81,82].

    CRH appears to be critical because HPA response to thedrug is blocked by peripheral administration of CRH butnot AVP antiserum [83]. Surprisingly, cocaine-inducedrelease of HPA hormones is not paralleled by enhancedc-fos expression in the PVN [76], although cocaineincreased CRH gene expression and lesions of the PVNmarkedly reduced the ACTH response to the drug [83]. Itwas suggested that cocaine may directly act at the PVNbecause the drug dose-dependently increased CRH releasefrom hypothalamic explants [84]. However, in pentobarbi-tal-anesthetized rats, local in vivo administration ofcocaine just posterior to the PVN requires doses muchhigher than i.c.v. administration to increase plasma corti-costerone, suggesting a main locus of action outside thePVN [82].

    HPA activation is found after local administration ofcocaine into the ventral striatum (that includes the nucleusaccumbens) [85]. However, there is no evidence for directprojections from the ventral striatum to the PVN and theprecise pathways are not known. Interestingly, CRH mayalso act within the brain to stimulate the HPA axis becausethe response was blocked by i.c.v. administration of a CRHantiserum and a CRH antagonist [86]. Dopamine appearsto be involved in cocaine-induced activation of the HPAaxis because peripheral administration of D1 and D2 re-ceptor antagonists partially blocked the corticosteroneresponse [75,8789] and reduced PVN CRH gene expres-sion caused by an acute cocaine binge [89]. A role fordopamine is also supported by the reduced response inDARPP-32-deficient mice to chronic administration ofcocaine [90] and blockade of the corticosterone responseto ventral striatum cocaine administration by haloperidol[85]. In addition to dopamine, other monoamines are likelyto be involved. Peripheral and i.c.v. administration ofdopamine, adrenergic (a and b), muscarinic and opioidreceptor antagonists has been found to reduce cocaine-induced activation of the HPA axis [75,87] and glutamateNMDA receptors also have a role [91,92]. Therefore, sev-eral different neurotransmitters participate in the acti-vation of the HPA axis caused by cocaine, suggesting amultifactorial regulation or a complex circuit whoseprimary place of activation is outside the hypothalamusand PVN.

    Methamphetamine (methylamphetamine), d-Amphet-amine, and 3,4-methylenedioximetamphetamine (MDMA,Ecstasy) exert complex biochemical effects on monoaminesynapses. They interfere with monoamine plasma andvesicular transporters, thus favoring the release of thethree monoamines from presynaptic terminals, indepen-dently of whether the synapses are active [93]. Amphet-

    Reviewamine has been found to dose-dependently increaseplasma corticosterone in rats [94,95], withmaximum levelsat 30-min post-injection. In our hands, HPA activation wasclearly observed at 90-min post-injection [96]. The mech-anisms involved in amphetamine-induced HPA activationhave been poorly characterized. HPA activation appears toinvolve CRH release because the response was blocked by aCRH antiserum [95]. However, we recently observed, usingdouble-labelling of c-fos and CRH, that amphetamine acti-vates a lower percentage of CRH neurons in the PVN thana severe stressor such as immobilization despite a similarnumber of activated neurons with both stimuli [97]. Thissuggests that other hypothalamic factors in addition toCRH may be involved in amphetamine-induced HPA acti-vation. Positive (but limited) evidence exists concerningmethamphetamine-induced activation of the HPA axis inexperimental animals [98]. At doses between 5 mg/kg and20 mg/kg, MDMA dose-dependently increased plasmacorticosterone in rats when given i.p., with a maximumat 1 h after administration [99]. It is of interest thatserotonin rather than catecholamines appears to beinvolved in the action of amphetamine, methamphetamineand MDMA on the HPA axis [94,99,100].

    ConclusionsThe data reviewed strongly suggest that all addictive drugsactivate the HPA axis acting mainly within the brain. Theprecise mechanisms and brain areas other than the PVNinvolved appear to greatly differ among the various drugs,and important information about events taking place atthe level of the PVN is lacking (see Table 1). Nevertheless,it is clear that classical classification of addictive drugs asstimulants or depressants does not contribute to explaintheir effects on theHPA axis, the particular mechanisms ofaction, or the development of tolerance after repeated drugadministration (not discussed here). Knowledge about howaddictive drugs activate the HPA axis is important for abetter understanding of the physiological and behavioralconsequences of acute and chronic exposure to drugs, andthe critical interaction between addictive drugs and stress.

    AcknowledgementsThis work was supported by grants SAF2008-01175 (MEC), PlanNacional sobre drogas, RD06/0001/0015 (Instituto de Salud Carlos III,Redes tematicas de Investigacion Cooperativa en Salud) and SGR2009-16. Thanks are given to David Rotllant, Xavier Belda and Raul Delgado-Morales for their help with preparation of the manuscript.

    References1 Koob, G. (2008) A role for brain stress systems in addiction.Neuron 59,1134

    2 Charmandari, E. et al. (2005) Endocrinology of the stress response.Annu. Rev. Physiol. 67, 259284

    3 Herman, J.P. et al. (2003) Central mechanisms of stress integration:hierarchical circuitry controlling hypothalamo-pituitary-adrenocortical responsiveness. Front. Neuroendocrinol. 24, 151180

    4 Armario, A. (2006) The hypothalamic-pituitary-adrenal axis: what canit tell us about stressors?CNSNeurol. Disord. Drug Targets 5, 485501

    5 Kovacs, K.J. and Sawchenko, P.E. (1996) Sequence of stress-inducedalterations in indices of synaptic and transcriptional activation inparvocellular neurosecretory neurons. J. Neurosci. 16, 262273

    6 Rivier, C. and Lee, S. (1996) Acute alcohol administration stimulatesthe activity of hypothalamic neurons that express corticotrophin-releasing factor and vasopressin. Brain Res. 726, 110

    7 Hauger, R.L. et al. (1988) Corticotropin-releasing factor receptors and

    Trends in Pharmacological Sciences Vol.31 No.7pituitary adrenal responses during immobilization stress.Endocrinology 123, 396405

    323

  • Review Trends in Pharmacological Sciences Vol.31 No.78 Vinson, G.P. et al. (1994) The neuroendocrinology of the adrenalcortex. J. Neuroendocrinol. 6, 235246

    9 De Kloet, E.R. et al. (1998) Brain corticosteroid receptor balance inhealth and disease. Endocr. Rev. 19, 269301

    10 De Kloet, E.R. et al. (2008) Corticosteroid hormones in the centralstress response: quick-and-slow. Front. Neuroendocrinol. 29, 268272

    11 Vuong, C. et al. (2010) The Effects of Opioids and Opioid Analogs onAnimal and Human Endocrine Systems. Endocr. Rev. 31, 98132

    12 Pfeiffer, A. and Herz, A. (1984) Endocrine actions of opioids. Horm.Metab. Res. 16, 386397

    13 Bartolome, M.B. and Kuhn, C.M. (1983) Endocrine effects ofmethadone in rats; acute effects in adults. Eur. J. Pharmacol. 95,231238

    14 Haracz, J.L. et al. (1981) Effect of intraventricular beta-endorphin andmorphine on hypothalamic-pituitary-adrenal activity and the releaseof pituitary beta-endorphin. Neuroendocrinology 33, 170175

    15 Pfeiffer, A. et al. (1985) Central kappa- and mu-opiate receptorsmediate ACTH-release in rats. Endocrinology 116, 26882690

    16 Iyengar, S. et al. (1987)Mu-, delta-, kappa- and epsilon-opioid receptormodulation of the hypothalamic-pituitary-adrenocortical (HPA) axis:subchronic tolerance studies of endogenous opioid peptides. BrainRes. 435, 220226

    17 Roy, S. et al. (2001) Role of hypothalamic-pituitary axis in morphine-induced alteration in thymic cell distribution using mu-opioidreceptor knockout mice. J. Neuroimmunol. 116, 147155

    18 Buckingham, J.C. (1982) Secretion of corticotrophin and itshypothalamic releasing factor in response to morphine and opioidpeptides. Neuroendocrinology 35, 111116

    19 Nikolarakis, K. et al. (1987) The role of CRF in the release of ACTH byopiate agonists and antagonists in rats. Brain Res. 421, 373376

    20 Lightman, S.L. and Young, W.S., 3rd (1988) Corticotrophin-releasingfactor, vasopressin and pro-opiomelanocortin mRNA responses tostress and opiates in the rat. J. Physiol. 403, 511523

    21 Zhou, Y. et al. (1999) Hypothalamic-pituitary-adrenal activity andpro-opiomelanocortin mRNA levels in the hypothalamus andpituitary of the rat are differentially modulated by acuteintermittent morphine with or without water restriction stress. JEndocrinol 163, 261267

    22 Laorden, M.L. et al. (2000) Activation of c-fos expression inhypothalamic nuclei by mu- and kappa-receptor agonists:correlation with catecholaminergic activity in the hypothalamicparaventricular nucleus. Endocrinology 141, 13661376

    23 Wang, X.Q. et al. (1996) Intracerebroventricular administration ofbeta-endorphin increases the expression of c-fos and of corticotropin-releasing factor messenger ribonucleic acid in the paraventricularnucleus of the rat. Brain Res. 707, 189195

    24 Reed, B. et al. (2008) Extracellular biotransformation of b-endorphin inrat striatum and cerebrospinal fluid. J. Neuroendocrinol. 20, 606616

    25 Mansour, A. et al. (1995) Opioid-receptor mRNA expression in the ratCNS: anatomical and functional implications. Trends Neurosci. 18,2229

    26 Jezova, D. et al. (1982) ACTH and corticosterone response to naloxoneand morphine in normal, hypophysectomized and dexamethasone-treated rats. Life Sci. 31, 307314

    27 Jezova, D. (1985) Stimulation of ACTH release by naloxone: central orperipheral action? Life Sci. 37, 10071013

    28 Siegel, R.A. et al. (1982) Effects of naloxone on basal and stress-induced ACTH and corticosterone secretion in the male ratsite andmechanism of action. Brain Res. 249, 103109

    29 Rivier, C. (1996) Alcohol stimulates ACTH secretion in the rat:mechanisms of action and interactions with other stimuli. AlcoholClin. Exp. Res. 20, 240254

    30 Richardson, H.N. et al. (2008) Alcohol self-administration acutelystimulates the hypothalamic-pituitary-adrenal axis, but alcoholdependence leads to a dampened neuroendocrine state. Eur. J.Neurosci. 28, 16411653

    31 Lee, S. et al. (2004) Site of action of acute alcohol administration instimulating the rat hypothalamic-pituitary-adrenal axis: comparisonbetween the effect of systemic and intracerebroventricular injection ofthis drug on pituitary and hypothalamic responses. Endocrinology145, 44704479

    32 Ogilvie, K.M. et al. (1998) Divergence in the expression of molecular

    markers of neuronal activation in the parvocellular paraventricular

    324nucleus of the hypothalamus evoked by alcohol administration viadifferent routes. J. Neurosci. 18, 43444352

    33 Quertemont, E. et al. (2005) The role of acetaldehyde in theneurobehavioral effects of ethanol: a comprehensive review ofanimal studies. Prog. Neurobiol. 75, 247274

    34 Kinoshita, H. et al. (2001) Acetaldehyde, a metabolite of ethanol,activates the hypothalamic-pituitary-adrenal axis in the rat.Alcohol Alcohol. 36, 5964

    35 Pastor, R. et al. (2004) Brain catalase activity inhibition as well asopioid receptor antagonism increases ethanol-induced HPA axisactivation. Alcohol Clin. Exp. Res. 28, 18981906

    36 Rivier, C. et al. (1984) Effect of ethanol on the hypothalamic-pituitary-adrenal axis in the rat: role of corticotropin-releasing factor (CRF). J.Pharmacol. Exp. Ther. 229, 127131

    37 Lee, S. et al. (2001) Mice that lack corticotropin-releasing factor (CRF)receptors type 1 show a blunted ACTH response to acute alcoholdespite up-regulated constitutive hypothalamic CRF gene expression.Alcohol Clin. Exp. Res. 25, 427433

    38 Lolait, S.J. et al. (2007) Attenuated stress response to acutelipopolysaccharide challenge and ethanol administration invasopressin V1b receptor knockout mice. J. Neuroendocrinol. 19,543551

    39 Ogilvie, K.M. et al. (1997) Role of arginine vasopressin andcorticotropin-releasing factor in mediating alcohol-inducedadrenocorticotropin and vasopressin secretion in male rats bearinglesions of the paraventricular nuclei. Brain Res. 744, 8395

    40 Li, Z. et al. (2005) Effect of ethanol on the regulation of corticotropin-releasing factor (CRF) gene expression.Mol.Cell.Neurosci.29, 345354

    41 Steiner, M.A. and Wotjak, C.T. (2008) Role of the endocannabinoidsystem in regulation of the hypothalamic-pituitary-adrenocorticalaxis. Prog. Brain Res. 170, 397432

    42 Weidenfeld, J. et al. (1994) Effect of the brain constituentanandamide, a cannabinoid receptor agonist, on the hypothalamo-pituitary-adrenal axis in the rat. Neuroendocrinology 59, 110112

    43 Manzanares, J. et al. (1999) Opioid and cannabinoid receptor-mediated regulation of the increase in adrenocorticotropin hormoneand corticosterone plasma concentrations induced by centraladministration of delta(9)-tetrahydrocannabinol in rats. Brain Res.839, 173179

    44 Puder, M. et al. (1982) Corticotrophin and corticosterone secretionfollowing delta 1-Tetrahydrocannabinol, in intact and inhypothalamic deafferentated male rats. Exp Brain Res 46, 8588

    45 Cota, D. et al. (2003) The endogenous cannabinoid system affectsenergy balance via central orexigenic drive and peripherallipogenesis. J. Clin. Invest. 112, 423431

    46 Saber-Tehrani, A. et al. (2010) Cannabinoids and their interactionswith diazepam on modulation of serum corticosterone concentrationin male mice. Neurochem. Res. 35, 6066

    47 Wenger, T. et al. (1997) Arachidonyl ethanolamide (anandamide)activates the parvocellular part of hypothalamic paraventricularnucleus. Biochem. Biophys. Res. Commun. 237, 724728

    48 Wenger, T. et al. (2003) The endogenous cannabinoid, anandamide,activates the hypothalamo-pituitary-adrenal axis in CB1 cannabinoidreceptor knockout mice. Neuroendocrinology 78, 294300

    49 Wade, M.R. et al. (2006) Cannabinoid CB1 receptor antagonismmodulates plasma corticosterone in rodents. Eur. J. Pharmacol.551, 162167

    50 Patel, S. et al. (2004) Endocannabinoid signaling negativelymodulates stress-induced activation of the hypothalamic-pituitary-adrenal axis. Endocrinology 145, 54315438

    51 Steiner, M.A. et al. (2008) Impaired cannabinoid receptor type 1signaling interferes with stress-coping behavior in mice.Pharmacogenomics J. 8, 196208

    52 Di, S. et al. (2003) Nongenomic glucocorticoid inhibition viaendocannabinoid release in the hypothalamus: a fast feedbackmechanism. J. Neurosci. 23, 48504857

    53 Ganon-Elazar, E. and Akirav, I. (2009) Cannabinoid receptoractivation in the basolateral amygdala blocks the effects of stresson the conditioning and extinction of inhibitory avoidance. J.Neurosci. 29, 1107811088

    54 Hill, M.N. et al. (2009) Suppression of amygdalar endocannabinoidsignaling by stress contributes to activation of the hypothalamic-

    pituitary-adrenal axis. Neuropsychopharmacology 34, 27332745

  • Review Trends in Pharmacological Sciences Vol.31 No.755 Rodriguez de Fonseca, F. et al. (1996) Corticotropin-releasing factor(CRF) antagonist [D-Phe12,Nle21, 38,C alpha MeLeu37]CRFattenuates the acute actions of the highly potent cannabinoidreceptor agonist HU-210 on defensive-withdrawal behavior in rats.J. Pharmacol. Exp. Ther. 276, 5664

    56 McLaughlin, R.J. et al. (2009) Monoaminergic neurotransmissioncontributes to cannabinoid-induced activation of the hypothalamic-pituitary-adrenal axis. Eur. J. Pharmacol. 624, 7176

    57 Cam, G.R. et al. (1979) The action of nicotine on the pituitary-adrenalcortical axis. Arch. Int. Pharmacodyn. Ther. 237, 4966

    58 Donny, E.C. et al. (2000) Differential effects of response-contingentand response-independent nicotine in rats. Eur. J. Pharmacol. 402,231240

    59 Chen, H. et al. (2008) Chronic nicotine self-administration augmentshypothalamic-pituitary-adrenal responses to mild acute stress.Neuropsychopharmacology 33, 721730

    60 Weidenfeld, J. et al. (1989) Further studies on the stimulatory actionof nicotine on adrenocortical function in the rat. Neuroendocrinology50, 132138

    61 Matta, S.H. et al. (1993) Nicotine stimulates the expression of cFosprotein in the parvocellular paraventricular nucleus and brainstemcatecholaminergic regions. Endocrinology 132, 21492156

    62 Matta, S.H. et al. (1997) Nicotinic activation of CRH neurons inextrahypothalamic regions of the rat brain. Endocrine 7, 245253

    63 Loughlin, S.E. et al. (2006) Nicotine modulation of stress-relatedpeptide neurons. J. Comp. Neurol. 497, 575588

    64 Matta, S.G. et al. (1998) Response of the hypothalamo-pituitary-adrenal axis to nicotine. Psychoneuroendocrinology 23, 103113

    65 Matta, S.H. et al. (1987) Nicotine elevates rat plasma ACTH by acentral mechanism. J. Pharmacol. Exp. Ther. 243, 217226

    66 Matta, S.H. et al. (1990) Role of the fourth cerebroventricle inmediating rat plasma ACTH responses to intravenous nicotine. J.Pharmacol. Exp. Ther. 252, 623630

    67 Matta, S.H. et al. (1993) selective administration of nicotine intocatecholaminergic regions of rat brainstem stimulatesadrenocorticotropin secretion. Endocrinology 133, 29352942

    68 Valentine, J.D. et al. (1996) Nicotine-induced cFos expression in thehypothalamic paraventricular nucleus is dependent on brainstemeffects: correlations with cFos in catecholaminergic andnoncatecholaminergic neurons in the nucleus tractus solitarius.Endocrinology 137, 622630

    69 Matta, S.H. et al. (1990) Catecholamines mediate nicotine-inducedadrenocorticotropin secretion via a-adrenergic receptors.Endocrinology 127, 16461655

    70 Fu, Y. et al. (1997) Adrenocorticotropin response and nicotine-inducednorepinephrine secretion in the rat paraventricular nucleus aremediated through brainstem receptors.Endocrinology 138, 19351943

    71 Fu, Y. et al. (1998) Nicotine-induced norepinephrine release in the ratamygdala and hippocampus is mediated through brainstem nicotiniccholinergic receptors. J. Pharmacol. Exp. Ther. 284, 11881196

    72 Zhao, R. et al. (2007) Nicotine-induced norepinephrine release inhypothalamic paraventricular nucleus and amygdala is mediatedby N-methyl-D-aspartate receptors and nitric oxide in the nucleustractus solitarius. J. Pharmacol. Exp. Ther. 320, 837844

    73 Okada, S. et al. (2003) Extrahypothalamic corticotropin-releasinghormone mediates (-)-nicotine-induced elevation of plasmacorticosterone in rats. Eur. J. Pharmacol. 473, 217223

    74 Rivier, C. and Vale, W. (1987) Cocaine stimulates adrenocorticotropin(ACTH) secretion through a corticotropin-releasing factor (CRF)-mediated mechanism. Brain Res. 422, 403406

    75 Sarnyai, Z. et al. (1993) Cocaine-induced elevation of plasmacorticosterone is mediated by different neurotransmitter systems inrats. Pharmacol. Biochem. Behav. 45, 209214

    76 Torres, G. and Rivier, C. (1992) Differential effects of intermittent orcontinuous exposure to cocaine on the hypothalamic-pituitary-adrenal axis and c-fos expression. Brain Res. 571, 204211

    77 Mantsch, J.R. et al. (2000) Effects of cocaine self-administration onplasma corticosterone and prolactin in rats. J. Pharmacol. Exp. Ther.294, 239247

    78 Mantsch, J.R. et al. (2003) Neuroendocrine alterations in a high-dose,extended-access rat self-administration model of escalating cocaineuse. Psychoneuroendocrinology 28, 83686279 Galici, R. et al. (2000) Comparison of noncontingent versus contingentcocaine administration on plasma corticosterone levels in rats.Eur. J.Pharmacol. 387, 5962

    80 Palamarchouk, V. et al. (2009) Self-administered and passive cocaineinfusions produce different effects on corticosterone concentrations inthe medial prefrontal cortex (MPC) of rats. Pharmacol. Biochem.Behav. 94, 163168

    81 Levy, A.D. et al. (1991) Cocaine-induced elevation of plasmaadrenocorticotropin hormone and corticosterone is mediated byserotonergic neurons. J. Pharmacol. Exp. Ther. 259, 495500

    82 Saphier, D. et al. (1993) Effects of intracerebroventricular andintrahypothalamic cocaine administration on adrenocorticalsecretion. Neuroendocrinology 57, 5462

    83 Rivier, C. and Lee, S. (1994) Stimulatory effect of cocaine on ACTHsecretion: role of the hypothalamus. Mol. Cell Neurosci. 5, 189195

    84 Calogero, A.E. et al. (1989) Cocaine stimulates rat hypothalamiccorticotropin-releasing hormone secretion in vitro. Brain Res. 505,711

    85 Ikemoto, S. and Goeders, N.E. (1998) Microinjections of dopamineagonists and cocaine elevate plasma corticosterone: dissociationeffects among the ventral and dorsal striatum and medialprefrontal cortex. Brain Res. 814, 171178

    86 Sarnyai, Z. et al. (1992) The cocaine-induced elevation of plasmacorticosterone is mediated by endogenous corticotropin-releasingfactor (CRF) in rats. Brain Res. 589, 154156

    87 Borowsky, B. and Kuhn, C.M. (1991) Monoamine mediation ofcocaine-induced hypothalamo-pituitary-adrenal activation. J.Pharmacol. Exp. Ther. 256, 204210

    88 Spangler, R. et al. (1997) Behavioral stereotypies induced by bingecocaine administration are independent of drug-induced increases incorticosterone levels. Behav. Brain Res. 86, 201204

    89 Zhou, Y. et al. (2004) Effects of selective D1- or D2-like dopaminereceptor antagonists with acute binge pattern cocaine oncorticotropin-releasing hormone and proopiomelanocortin mRNAlevels in the hypothalamus. Brain Res. Mol. Brain Res. 130, 6167

    90 Zhou, Y. et al. (1999) Effects of chronic Binge cocaine administrationon plasma ACTH and corticosterone levels in mice deficient inDARPP-32. Neuroendocrinology 70, 196199

    91 Torres, G. et al. (1994) A ketamine mixture anesthetic inhibitsneuroendocrine and behavioral consequences of cocaineadministration. Brain Res. 656, 3342

    92 Damianopoulos, E.N. and Carey, R.J. (1995) Evidence for N-methyl-D-aspartate receptor mediation of cocaine induced corticosteronerelease and cocaine conditioned stimulant effects. Behav. BrainRes. 68, 219228

    93 Elliott, J.M. and Beveridge, T.J. (2005) Psychostimulants andmonoamine transporters: upsetting the balance. Curr. Opin.Pharmacol. 5, 94100

    94 Knych, E.T. and Eisenberg, R.M. (1979) Effect of amphetamine onplasma corticosterone in the conscious rat. Neuroendocrinology 29,110118

    95 Swerdlow, N.R. et al. (1993) Pituitary-adrenal axis responses to acuteamphetamine in the rat. Pharmacol. Biochem. Behav. 45, 629637

    96 Gagliano, H. et al. (2009) Repeated amphetamine administration inrats revealed consistency across days and a complete dissociationbetween locomotor and hypothalamic-pituitary-adrenal axis effects ofthe drug. Psychopharmacology (Berl) 207, 447459

    97 Rotllant, D. et al. (2007) Differential effects of stress andamphetamine administration on Fos-like protein expression incorticotropin releasing factor-neurons of the rat brain. Dev.Neurobiol. 67, 702714

    98 Williams, M.T. et al. (2006) Ontogeny of the adrenal response to (+)-methamphetamine in neonatal rats: the effect of prior drug exposure.Stress 9, 153163

    99 Nash, J.F. et al. (1988) Elevation of serumprolactin and corticosteroneconcentrations in the rat after the administration of 3,4-methylenedioxymethamphetamine. J. Pharmacol. Exp. Ther. 245,873879

    100 Doyle, J.R. and Yamamoto, B.K. (2009) Serotonin 2 receptormodulation of hyperthermia, corticosterone, and hippocampalserotonin depletions following serial exposure to chronic stress andmethamphetamine. Psychoneuroendocrinology [Epub ahead of print]325

    Activation of the hypothalamic-pituitary-adrenal axis by addictive drugs: different pathways, common outcomeIntroductionOpiates and eOPsEthanolCBsNicotineCocaine and amphetamine-like drugsConclusionsAcknowledgementsReferences