Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. ...HmnStudies Human 2.1.1. reward. Outcomes and Addiction-Related analgesia androgens Effects 2.1 how Opioid-Related of and is in areas review effects, Differences effects, in this opioid-mediated Sex opioid-mediated of especially on goal differences, in 2. androgens The sex androgens of relevant literature. of effects clinically this role the to synthesize contribute the to examining may attempts research & described on comprehensive Berry, the has effects any Huhn, describe of literature their 2020; aware to of described Perotti, in not have & body are progesterone, Kokane we articles smaller and and 2009; review A estrogen Fillingim, of of particularly 2016; 2018). number much hormones, Koob, , Dunn, a ovarian and to and of (Becker regard effects, outcomes role In deter- opioid-related opioid-mediated the classes. in opioid- of on pharmacological role in modulation focused many known hormones the from their has given gonadal drugs research investigation of of prior for efficacy role the target and the obvious potency concerns an the are attention mining hormones research Chartoff, explored. Gonadal and significant been & analysis, prescribed received effects. have Becker of mediated women has differences levels 2020; that these and epidemiological to Perotti, area reward men and contribute One & opioid-mediated clinical, in may Kokane preclinical, that in disorder 2020; at mechanisms differences use apparent of al, sex number are opioid et et a Similarly, differences Neisters an (Lopresti sex 2008; developing recreationally These 2013). (Craft, of opioids 2019). Ho, pain risk using chronic & the or and efficacy Lee in opioids acute and 2014; differences for Murphy, potency to treated the & contributes women in Loyd and differences differences 2010; sex of men significant instance, Many al., between to For effects. analgesics contribute implications. opioid-mediated opioid analgesia health in of opioid-mediated public differences have to gender and sensitivity and meaningful in sex clinically significant are reveals differences literature these of body large A Introduction conditions 1. addressing that for modulation conclude options androgenic We new disorders. investigators, of to use clinical lead substance implications and may and clinical preclinical effects pain the chronic opioid-mediated for as of research addresses such modulation of also androgenic lines review into future contribute investigation The may suggests further androgens and how effects. behaviors and ovarian opioid-mediated endpoints opioid-mediated describes of opioid-mediated review of in on role This androgens differences the androgens. of examined sex of influence has role the to research mediated the regarding much are examined known Whereas differences has currently research system. is sex less opioid what these comparatively endogenous of endpoints, the Many opioid-mediated on functional on females. to hormones hormones leading and gonadal receptors, males of opioid between effects and the behaviors peptides opioid by opioid-mediated endogenous to in sensitivity differences sex in are differences there indicate data of abundance An Abstract 2020 2, October 1 Androgens of Sharp Jessica Role Effects: Opioid-Mediated in Differences Sex aisnCollege Davidson 1 alaPearson Tallia , 1 n akSmith Mark and , 1 1 Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. ffcso ap pod.Frisac,ml asaqieaU953v.sln iciiainfse than faster discrimination saline vs. U69,593 al., a stimulus et discriminative acquire (Craft the rats to apparent male sensitive longer more instance, no are For rats are male opioids reinforcement opioids, opioids. mu of eliminate kappa 1996). with to effects of seen al., changed stimulus that effects et complicate are to discriminative contrast parameters can (Craft the In schedule which vehicle tests in 1998a). when and females, differences drug substitution Indeed, than between sex in frequency opioids sexes. mu reinforcement bias, of females two in of females, biases the effects than to than effects between rate-suppressing due males rate stimulus sessions the data slower in discrimination discriminative to a drug potent the sensitive at of interpretation more discrimination to less saline are sensitive are rats vs. less the agonists Male both be opioid a to may acquire mu sensitive rats rats and less male male also example, are that For rats rats evidence opioids. male Male some that such is females suggesting 2017). factors opioids. than There by 2008), of al., doses vary effects Zarrindast, et of can rewarding & (Mavrikaki range conditioned effect Karami narrower restriction and this a 2000; reinforcing food but over al., 1999), of to preference et 2003; Carroll, place level longer al., (Cicero & conditioned take and et Lynch morphine-induced and Cicero reinforcement, a intake 1978; 2002; of 2020; demonstrate al., opioid al., schedule Perotti, et et of (Carroll & dose, levels Alexander Kokane females lower as 2016; to 2008; exhibit al., compared and Craft, rats self-administration et 2016; opioid male of Lacy Koob, IV effects instance, & and reinforcing For Becker oral and by acquire rewarding 2019). reviews the Chartoff, (see to & rats sensitive Becker less female are than rats opioids male conditions. other that control indicate time. relevant studies this lack Most 2012; at and unknown al., sizes mostly Studies et a is sample Animal (Blick are intake small opioid users There 2.1.2. have in opioid studies androgens humans. in of these in affect role but that effects the positive 2009), Consequently, studies abuse-related in experimental Zylicz, in any increases & find androgens sex testosterone-induced Roantree not on of describing could hormones role reports we gonadal potential and few of Ho, the populations, and influence & human possible addressed stress, in Lee the 2018). of systematically use/abuse al., examining levels 2011; opioid et studies greater al., in (Moran few with differences very men et Kokane then been associated Back 2007; have situations al., are 2007; there stressful et consequences Remarkably, Greenfield during al., negative see craving et review, These opioid (Yu (for Giacomuzzi more lifetime craving men. 2020; experience their women than more Perotti, over treatment Bawor 2020) & experience seek 2013; (Kokane Perotti, physical 2018), to al., symptoms & likely more et al., withdrawal less report McHugh more are et 2011; 2015), and report Dunn al., 2013), al., 2019), et 2005, et al., Chartoff, Back Kaplovich al., & et 2003; see Becker et al., Huhn 2011; but et 2015a; al., 2020; (Cepeda et al., al., effects Back et side see et Hser, negative review, Lopresti, 1987; (for socioeconomic McGlothin, 2020; faster and & For Perotti, doses Hser, & men. higher (Anglin, Kokane to to quicker compared see escalate 2019; addiction use 1897), 2017; for opioid Booth, to criteria al., & related diagnostic Anglin, et consequences the Serdarevic negative meet disorder experience 2009; women use 2020). to example, al., likely substance NIDA, more a et 2012; also have Green al., are and Women 2020; et 2020). abuse (NIDA, Darnell Perotti, opioid & men 2003; for from Kokane than Carr, suffer criteria by to diagnostic opioids & review likely may meet prescription Cepeda more findings to to are 2009; These likely and attributed al., 2020). more pain opioids, et al., also bodily prescribed (Fillingim et general are be (Schieber more Women men to report men than likely et women than (Bawor more disorders that filled are opioids observation pain 2020), prescriptions prescription the Perotti, opioid with to & have 2011; related experience Kokane to al., be opioid 2013; et likely Ho, first 2011; (Back more & al., their opioids are in Lee et have prescription by and heroin Back to use reviews 2010; use likely to see al., heroin, more likely et 2015a; use are more (Back al., to women are 2009), likely than women sex al., more time contrast, notable et overdose are of In also Green men of period are 2018 2020). instance, risk longer There NIDA, For in a the 2016; 2020). for opioids. overdose, States Kuhn, al., heroin illicit opioid et use United vs. an (Lopresti and licit quantities, the 2000 experience of larger since to in use men likely the overdoses than more in women d 2 Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. diitrdvaspapnlta pnlo eihrlrue seCat 08 aa ta. 08 onr& are Bodnar opioids 2008; when al., therapeutic apparent et a more Dahan are 2008; across Craft, differences males (see These in routes antinociceptive peripheral 2010). efficacy or greater Kest, and/or spinal in & produce than potency sensitivity Bodnar supraspinal agonists greater via 2008; nociceptive opioid by administered Craft, models, baseline evidenced (See these greater females, range In exhibit than dose males 2020). animals in (Mogil, female effects pain humans, of with models observed 2004). preclinical Beckham, that & opioid to Zacny to 2004; Similar sensitive al., more differences et are sex Studies (Fillingim men measuring measures Animal that studies pain Fillingim 2.2.2. report Laboratory all) 2004; generally not 2004). al., (but but Beckham, et some equivocal, & (Al’Absi lower on more have Zacny measures analgesia women are 2005; Similarly, pain analgesia al., laboratory 2002). opioid-induced Niesters pain et al., on in et alleviate 2020; Fillingim men Chia to Mogil, 2004; see than morphine al., but 2014; tolerances more Murphy, 2003; et pain significantly Carr, & and & require Loyd in Cepeda thresholds and 2013; analgesia 2005; several pain pain al., Ho, and opioid more et 2020), & to (Aubrun report men Mogil, Lee sensitivity women than Clinically, 2019; 2009; and Afify, 2010;). sensitivity al., & al., et pain Rasakham Nasser et 2010; Fillingim in 2014; al., Murphy, 2008; differences et analgesia/antinociception & (Craft, sex Niesters opioid Loyd 2010; humans addressed Kest, 2013; in specifically & Ho, differences have Bodnar & 2009; sex reviews Lee al., on et 2011; Fillingim published Liu-Chen, 2008; & been al., et have Dahan reviews 2008; excellent (Craft, of number A Studies Human et (Craft 2.2.1. females Analgesia/Antinociception and Opioid-Induced males and opioid-induced Pain between the equivalent in 2.2 if functionally 2002a). differences especially al., not sensitivity, sex et opioid is tolerance, Cicero acute 2008). administration opioid-induced see in al., chronic but differences al., in for 2009; sex et differences chosen al., consideration (Craft sex into dose et take shakes to Sagedhi must 2008; wet-dog regard dependence Rezazadeh, and in physical & jumping, above Nayebi with- 2005; -precipitated loss, noted severe 2001, weight As more al., greater display withdrawal et (Cicero also including Diaz species earlier rodents 1999; females, across Male and vary than spontaneous 2006). symptoms scores, undergoing symptoms Contarino, withdrawal withdrawal & rodents drawal of Papelo Male higher duration 2002a; the 2010). al., loss, although re- et Kest, weight females, (see & to greater withdrawal Bodnar compared exhibit and 2016; this symptoms dependence Koob, morphine Supporting physical & from Becker opioid-induced 2008). withdrawal 2008; for al., Craft, reported 2001). by et been al., views have et (Dahan females, (Barrett differences females functionally in chronically sex when than are than administered tolerance Similar males males are males opioid-induced in If drug of in greater a degrees functionally 2.2.2). dose comparable of is Section a demonstrate doses that dose rats (see equivalent that that at female basis meaning if and greater mg/kg and opioids, male be a opioid, of notion, an on will effects of acute females antinociceptive tolerance dose the than acute given then to males a sensitivity the in to in to sensitive greater differences sensitive more sex functionally more to is are part, dose Males in given relative due, opioids. males be may of in 2001). tolerance development effects al., tolerance opioid-induced tolerance et which in of (South differences to rate females but Sex extent testosterone-treated the 2007; the and reduces al., in males castration et For differences intact Mousavi and hormones. sex both 2001; gonadal 2007), abolishes to by al., al., part, females et 1984; in et and Barrett George, mediated, (Mousavi 1999; are males & develops differences al., both Kasson sex et 2005; These of (Craft Wala, gonadectomy females. extent & than instance, greater 2004) (Holtman al., a rate et to faster Holtman and see a 2001) at al., differences tolerance sex et Furthermore, kappa develop consistent South which rats of in Male effects areas tests. additional rate-suppressing observed. two substitution are are the dependence in to physical observed and females tolerance not Opioid-induced than are 1998b). males differences al., sex in et agonists, potent (Craft mu agonists more with seen are that agonists unlike kappa and females, 3 Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. omnsi eae eg,Foe ta. 03.Eauto fteesxdffrne sas opiae by complicated sexes. also across is and Peptides differences within Opioid both sex Endogenous vary these 2.4.1. that of roles system ovarian regulatory Evaluation opioid circulating multiple endogenous of suggesting 2003). stages, the function al., developmental of a and et as regions wanes neuroanatomical Flores across and (e.g., inconsistences waxes differences is females sex systematically these (see there magnitude in Evaluating females 2015). and the Mavrikaki, and hormones because & 1995), males Chartoff between difficult Reisine, 2011; differ often Liu-Chen, 2012; receptors & is and Hallberg, Rasakham peptides 2018; & al., these et (Nyberg of to Huhn concentrations respectively bind the receptors, that preferentially evidence opioid ample kappa and and met/leu- delta beta-endorphin, mu, peptides, opioid al., endogenous Peptides et The Opioid 1984). (Kest George, and morphine & 2003; Receptors of Kasson al., effects Opioid less see et hyperthermic) 2.4 are but rodents (Elliott and 1985; male hypothermic morphine al., contrast, of (both et In Quock thermoregulatory 1999). effects latter 2000; al., the immunological/inflammatory this et to the although Sarton and rodents, to 2008; 2000) female 2006) al., rodents al., than et female et Dahan morphine (Craft than mice (see of antidiuretic sensitive female species 1999) the than across al., to vary morphine et sensitive may of (Craft more effect effects effects are Rodaros, depressant locomotor-stimulating & rodents Stewart respiratory the male 2004; Moreover, and to al., 1986). sensitive et Innes, Holtman more 2006; & locomotor- are (Kavaliers al., the mice et to male sensitive (Craft gene are more rats and immediate-early rats are female 1999), rats than the male male morphine example, of Similarly, of For 2002). expression effects al., effects. suppressive morphine-induced et drug (D’Souza to tail unconditioned rats Straub to morphine-induced female extend than often sensitive sensitivity more opioid in differences Sex Opioids al., of et Effects Terner et 2003; Unconditioned (Mitrovic al., 2.3. mice et hormonal 3.2.2). Elliott wildtype via (e.g., Section in manipulated androgens see apparent be endogenous and are can of 2002, that antinociception perturbation differences opioid-induced via sex particularly in the manipulation, differences exhibit sex GIRK in- Alternatively, not lack 2002). (Holtman to do generally na¨ıveal., that subjects leads and in manipulations observed mice mice, administration those difference mutant wildtype morphine sex Similarly, chronic than but the instance, 2004). abolishes manipulations, For al., and of sexes. sexes et number both both in in a tolerance effective by antinociceptive less eliminated opioids rendering be among volve than can analgesia commonality opioid-induced obvious differences to no sensitive females Sex is more in there al., be sensitivity 2000), et settings. would greater al., women Stoffel therapeutic which of et al., 2004; in in Tershner examples et Wala, men conditions 2005; Barrett some & predict al., 2000; Although Holtman to al., et 2004; studies 2011). et Stoffel these al., al., Cook (e.g., et et 1999; et reported Holtman Peckham al., Bai been 2002; 2005; et have al., Craft al., mechanical: et 1996; et (e.g., Terner al., Cataldo these conditions 2001; et 2005; experimental Cicero but Bernal, & of thermal: thermal), Craft 1996; majority males 2001; al., the vs. with et under mechanical Cicero endpoints, females 2015; experimental (e.g., al., than across stimuli differences sensitive differences Finally, nociceptive qualitative more 2008). across 2008). than being al., (or vary al., rather et quantitative amplify et can (White involve to Dahan groups females typically differences 2010; age three also and sex across Kest, the differences males females with & – sex and interact between at Bodnar these males can efficacy see between of intrinsic antinociception review, differences magnitude relative opioid-induced for minimize) The their George,1984; in delta; and & differences 2006). kappa, – age Kasson 2003; mu, for Moreover, al., 2002; (i.e., selectivity et relative receptors al., differ- Terner their opioid et large 2000; on primary (Barrett to al., based minimal females et opioids from and across Cook ranging vary males 1999; subject, between al., of efficacy et strain Kest and/or across potency differs differences in sex ences pharmacodynamics females. these the and of in males magnitude differences between The by drugs primarily these mediated of are pharmacokinetics) differences than sex (rather that suggesting 2010), Kest, 4 2 r essniiet podidcdantinociception opioid-induced to sensitive less are c-Fos n to and Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. et eetr nC3prmdlclso h ipcmu,btti ffc sas eedn nteestrous the on dependent also is effect this of but sex concentrations cases, hippocampus, greater these the exhibit of Females of cycle. both cells et estrous In pyramidal (Williams the 2002). CA3 females of McDonald, in phase in also & receptors proestrus effect cycle Mascagni, delta this the (Wilson, estrous but to females the amygdala, confined of the of are in phase differences phase concentrations CA1 estrous the receptor the the delta on within on than greater concentrations dependent have depends gyrus receptor rats is delta dentate male this Similarly, greater the 2011). but have within al., also hippocampus, rats receptors the Male opioid of 2011). delta region 2003). al., of et al., (Williams concentrations et rats (Morely-Fletcher greater female region significantly brain have across feminizing and rats and varies sex Male masculinizing the too producing by effect that fetuses minimized this female hypothalamus matters or however, and exaggerated the Complicating Data male respectively; be 1991). nearby in 2008). effects, either with al., al., findings siblings, can et et intrauterine concentrations conflicting Maggi of Loyd receptor 1991; with position mu F; al., in clear, > et differences functional (M less Limonta sex to area are further, 1995; traced Vathy, grey (Riman´oczy be areas & periaqueductal age can and other across the F) 2007), in vary > in al., (M et significantly differences receptors Bernal opioids have mu sex 2008; mu of rats examining al., of density Male et effects the (Kren antinociceptive subtypes. midbrain in the receptor and differences to opioid cord sensitivity spinal in major the differences three in sex levels all protein for receptor apparent mu higher also are differences Sex Receptors spinal Opioid both 2000). the to 2.4.2. in al., relative (Brads- levels et estrus cord preprodynorphin (Bradshaw in spinal decreases males rats the significantly female of hormones. in castration and cord levels and gonadal rats males, preprodynorphin of male gonadectomized levels increases activity both ovariectomy and mRNA the to Moreover, intact preprodynorphin relative by 2000). males. CFA higher part al, to of significantly et large injection compared have haw following in 2013) proestrus cord due al., and spinal are diestrus et the differences in within Kempen sex rats (Van females these hippocampus instance, that the dynor- For evidence greater of also and region is 1991), CA3 (Simerly, brain containing There to hypothalamus the neurons critical compared the in more pituitary few of concentrations significantly nucleus anterior a phin have periventricular the anteroventral females across in the whereas concentrations in dynorphin 1986), dynorphin mRNA of al., in concentrations et differences higher (Molineaux have sex females males suggests instance, data For of regions. of amount signi- nucleus limited have ovariectomy periventricular 1991). males A anteroventral and (Simerly, Finally, the males females 1983). within in to Hong, compared castration mRNA & (Yoshikawa hypothalamus both differences the containing that these given neurons These of hormones, more 1991). magnitude gonadal ficantly Man, the to & pituitary, reduces signi- Tang part, anterior have females 1983; the in males the Hong, in in due, adult including & Yoshikawa are leu-enkephalin pituitary, contrast, 1982; differences In the and Lamartinere, males. sex in 1991) & Yoshikawa, than leu-enkephalin Man, (Hong, 2013) and females & al., met- than (Tang et concen- of cortex Kempen greater concentrations (Van cerebral and greater hippocampus 1986) ficantly the the al, in concentrations of et leu-enkephalin greater region (Watson in have and CA3 area emerge females met- preoptic differences adult the of sex instance, within For trations significant fibers specific. Some immunoreactive region 1979). met-enkephalin are al., of differences telencepha- et these met/leu- (Bayon diencephalon, in and amygdala midbrain, differences adulthood, and medulla, sex striatum, the exhibit not including hippocampus, do regions, lon, pups many rat across prepubescent concentrations beta-endorphin, enkephalin for reported that to males Similar than 1986). pituitary Wardlaw, anterior 2009; and al., pituitary hippocampus exhibiting et hypothalamus, females exhibiting the adult males (Pluchino the areas, with adult of in and key with concentrations females, areas, lobe several than some beta-endorphin neurointermediate cortex in in greater prefrontal emerge the the adulthood differences in Sex and concentrations into 2009). beta-endorphin continues al., lobe, greater et levels Pluchino parietal 1982; beta-endorphin et brainstem, al., the in et (Bayon eminence, (Petraglia in differences amygdala median beta-endorphin sex and the of of cortex, concentrations including lack striatum, in A differences hippocampus, 5 Posted on Authorea 2 Oct 2020 | The copyright holder is the author/funder. All rights reserved. No reuse without permission. | https://doi.org/10.22541/au.160167496.62768848 | This a preprint and has not been peer reviewed. Data may be preliminary. h podatgns ateoebok h enocn ffcso etseoei natml ases(Peters hamsters male intact with in positive shell pretreatment testosterone the accumbens that Importantly, of given nucleus 1999). effects activity, reinforcing al., the et opioid the et into blocks endogenous Arnedo (King injections by area 1994; antagonist mediated preoptic site-specific opioid al., are medial the or or testosterone et 1996) 1997) of (Alexander al., al., place effects et reinforcing injections et Packard conditioned 2002; Caladrone systemic a al., see its either et produces of (Frye but following Testosterone doses 2002; rodents high) 2006). 2000, male not al., Wood, (but intact self-administer & low in also circulating (DiMeo and preference will that (DHT), estradiol suggesting hamsters dihydrotestosterone metabolite, 2004), testosterone, Male antiandrogenic Wood, of self-administration. & metabolite intact testosterone (DiMeo androgenic in to the hamsters testosterone sensitivity male of intake increase intraventricular castrated infusions increases intraventricular androgens in replacement of Testosterone and testosterone infusions 2004). 2004) intravenous exogenous Wood, by al., & of maintained (Peters et hamsters reliably (Wood female be and rats can male male Intact choice poking two-bottle intact nose models. a Similarly, in in preclinical vehicle 2001). testosterone than Wood, in testosterone & oral effects (Johnson of rewarding amounts test greater conditioned self-administer reliably and hamsters reinforcing male positive produces 1999). liability Testosterone al., abuse men. et the in Kindlundh true enhance 1996; with particularly mutually al., Animals be may concurrently et 3.1.2 may AAS McBride and 2010; this and 2009; to and Rane, (G˚arevik al., agonists another, & abuse prior et opioid one Skarberg AAS that occurs of 2009a; 2014; to suggest also al., al., susceptibility findings et use et increases such addiction Ranjan Collectively, Kanayama Opioid also opioid 1999; of use 2005; 2003). al., risk et opioid Currier, al., the Wines that & increases et suggesting (Trenton abuse Kanayama abuse, AAS use 2018; that AAS et AAS conclude al., admission Kanayama to of between et investigators 2009; relationship history leading (Kanayama al., a 2014), prior et reported al., to a Skarberg have et precursor studies 2003; and Ranjan a several al., treatment often Indeed, et is opioid 2009b). abuse (Kanayama al., for AAS opioids et humans, particularly Kanayama females In 2009a; to drugs, 2008). compared al., Wood, other (AAS) 2018; of steroids al., abuse anabolic et androgenic the Kanayama abuse 2010; and al., use et to (Kanayama likely more significantly are Males Humans 3.1.1 Outcomes Abuse-Related Chang, of Effects 3.1 phase Harris, Opioid-Related the 2007; of account al., into Control et available. take Androgenic Drake are should 3. 2011; sensitivity data these opioid those al., in when that et differences cycle Williams evidence 2000; sex estrous 2008; Given al., of Murphy, the et fashion. determinations & Bradshaw 2004), Wang, cyclic (e.g., Drake, Loyd, a the consequences In & 2003; functional of in sexes. al., have course differences both concentrations et the sex receptor in Flores over and eliminating hormones significantly peptide vary then gonadal fluctuate in receptors and by variations receptors opioid creating regulation and and dynamic cycle, peptides of under estrous peptides endogenous concentrations are Importantly, of opioid stage. proteins concentrations in developmental females, and differences and regarding peptides age that known various across reveal vary is these section and Less region 2004). this neuroanatomical al., in across et described both Harris forebrain. in studies 2007; the increasing al., The within and et with receptors (Drake proestrus cycle, kappa estrus during estrous in cord during the (Kren, differences on spinal medulla males sex dependent the and of to highly within cord compared concentrations are increasing spinal differences midbrain) females higher the These necessarily in significantly 2007). not density al., have (but receptor et kappa hindbrain rats Drake and 2008; female Welch, cord & that spinal Haller, suggest the in data memory 2016). receptors available and kappa al., learning the et in (Mazid limited, differences stressors sex Although delta of for alter types implications stress different has chronic to which and response fashion, acute dimorphic further, in sexually matters a Complicating in proestrus. receptors during apparent only and cycle 6 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. ta. 07 aai ta. 05 liie l,21;Dnele l,20;Aiiaie l,21;Roantree al., 2019; et female al., (Aloisi from et testosterone transitioning with AminiLari treatment individuals 2006; upon transgender al., sensitivity Finally, et pain 2020). Daniell in al., decrease 2011; a et al., (Raheem demonstrate Glintborg baselines et typically 2010). own see pain Aloisi male al., their but to 2015; increases et or Teepker 2009, al., therapy controls 2015; Zylicz, et placebo replacement al., & Basaria either et testosterone to 2017; (Bartley relative that al., men sensitivity et report in pain pain are hypogonadism decreases greater they and with with as thresholds associated men associated just positively inversely women, examining are in testosterone are Studies of thresholds estradiol levels pain circulating of however, testosterone greater 2015); levels receiving al., with hypogonadism et Circulating (Bartley with women 2020). in men in decreases and al., thresholds (i.e., 2018) et thresholds al., pain et (Glintborg (Apkhazava greater therapy men with normal associated in positively sensitivity) are pain testosterone free of levels 2005) Circulating Currier, & overlapping Trenton Studies some 1989; Human Kleber, suggesting 3.2.1. & withdrawal, Activity Kashkin Androgenic opioid 1989; and al., of et Antinociception those Brower 3.2. (see to play similar at qualitatively be symptoms the may are of mechanisms many treated withdrawal However, or mice 2004). 2001), AAS Wood, male & al., of (Peters intact et testosterone (Negus in self-administering testosterone withdrawal hamsters with male precipitate treated agonist intact monkeys mu not male concurrent intact do of (C´el´erier 2003), absence nandrolone antagonists al., with the opioid et in instance, syndrome withdrawal For opioid-mediated administration. an naloxone-precipitated 2007). produce decreases Ahmadiani, not finasteride & do (Verdi with Sadeghi Androgens treatment rats 2008; chronic male Rezazadeh, and morphine-treated & acute (Nayebi in both testosterone withdrawal contrast, with opioid In treatment supplemental 2009). rodents, antagonist, by al., male receptor blocked et morphine-treated is androgen in effect the withdrawal this naloxone-precipitated Rezazadeh, and with physical & attenuates (Nayebi opioid-induced treatment castration rats et of Similarly, premise, male severity morphine-treated 2008). Sadeghi this the in 2008; withdrawal increase for naloxone-precipitated Rezazadeh, androgens attenuates support flutamide, that & further suggesting in Nayebi Lending 2003), withdrawal 2003; al., naloxone-precipitated dependence. (C´el´erier et al., of Philipova rodents et severity 2009; female the al., and increases in male generally morphine androgens morphine-treated of with effects antinociceptive treatment mice 2007). the Chronic Ahmadiani, male to & tolerance intact (Verdi of rats in development male the 5 morphine intact attenuates chronic a Similarly, of DHT, finasteride, 2003). to effects with al., testosterone et treatment antinociceptive (Philipova acute) rats the not female and to (but male nandrolone tolerance gonadectomized with in treatment of and 1999). chronic (C´el´erier 2003) instance, development al., For al., et opioids. et the in to (Craft increases tolerance attenuates castration by of following development evidenced the opioids as morphine decrease mu tests, for Androgens of discrimination substitute effects androgens drug to endogenous rate-suppressing in Paradoxically, agonists the opioids 1999). mu mu to opioid al., of of sensitivity mu et effects efficacy (Craft of rate-decreasing castration and effects the following potency stimulus attenuate assay the opioids discrimination discriminative mu drug in the of a reductions enhance effects al., in by stimulus androgens et discriminative evidenced (Wood endogenous the as modulate models instance, androgens agonists, animal For but traditional rats. 1992), in al., male stimulus et 2014). in Beun discriminative Wood, De a & see reinforcement as (Cooper but of serve 2011; schedule schedule reliably PR FR1 not a nandrolone an does on Testosterone with on intake treatment responding morphine rats alter chronic male not Moreover, morphine-induced intact does 2003). reduces in (C´el´erierbut but al., intake mice 2016) morphine et al., male decreases et intact significantly across (Chow enhan- vary rats in nandrolone, opioids male agonist, preference intact of receptor place in effects androgen preference rewarding the place with conditioned morphine-induced Pretreatment and ces reinforcement. reinforcing of positive schedule the and on species androgens of effects The 2004). Wood, & α- euts nyeihbtrta rvnsterdcinof reduction the prevents that inhibitor enzyme reductase 7 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. steeidct htadoescnitrc ihfcossc sseis podrcpo utp,and subtype, receptor opioid species, such as Findings such the 2005). factors reduces al., testosterone with processes. et antinociceptive and (Stoffel interact opioid-mediated 2016), rats can influence al., male to androgens et gonadectomized assay that Tsutsui behavioral in effects 2003; indicate agonist decreases antinociceptive al., these delta androgen-induced the et as a of (Philipova reduce of rats reports not DHT effects male isolated does and antinociceptive intact by also nandrolone in treatment complicated example, Testosterone morphine also For 1998). of ovariectomy is following al., antinociception. picture monkeys et opioid-mediated female The monkeys (Cook in in 2002). male opioids sheep Mello, in mixed-action in or & morphine kappa, agonist (Negus of mu, kappa effects effects a antinociceptive 2003), antinociceptive (C´el´erier or the al., the mice alter 2001) testosterone receptor et alter male by al., androgen not in or et the does morphine 1999) to (Negus treatment of Bodnar, exposure testosterone effects & chronic Krzanowska chronic antinociceptive nor 1989; and the acute al., influences neither 1996; et Similarly, al., nandrolone, Kepler et 2011). agonist, (Cicero in 1993; al., castration al., studies by et multiple et altered (Peckham not Islam instance, replacement are For 2002b; several morphine sensitivity. al., are of opioid effects et antinociception antinociceptive on Cicero opioid-mediated the testosterone manipulation that in androgen report increases androgens rats of finasteride of male effect Finally, role no the 2007). reporting of Ahmadiani, 2006). follow- studies & understanding rats al., (Verdi across clear male Moreover, et morphine observed a adult of Sumner 2005). Complicating are in effects al., 2005; effects antinociceptive agonists et the these 2003, kappa decreases Stoffel enhances and and al., castration 2002; and mu rats, al., et instance, concentrations both et adult (Stoffel For Terner of in castration 2015; effects agonists rats. ing al., antinociceptive kappa et adult the (Bai and increases in assays mu testosterone reported behavioral previously of and been described range strains antinociception have wide different et opioid effects Krzanowska a in 2002b, Similar to differences al., sex sensitivity et 2.2.2). of (Cicero magnitude neonatal Section adulthood in the (See in treatment reducing testosterone morphine thereby addition, androgens of nocicep- 2002), In that effects inflammation-related al., 2002). suggest antinociceptive and al., thermal data the et both of increases Krzanowska in 2006; abundance females Fuchs, rats an & male but (Borzan adult decreases assays in 2020), castration tive morphine neonatal Afify; of et instance, & effects For Craft antinociceptive Nasser by conditions. the and reviews many under (see 2008; antinociception equivocal opioid-mediated al. somewhat increase et are Dahan antinociception opioid-induced 2004; on al., androgens 2019). thresholds of nociceptive al., effects on et The swimming testosterone (Sharma Finally, of administration. opioids effects testosterone 2005). endogenous by the 2004, by enhanced that Frye, mediated is suggesting & is effect effect, (Edinger this this rats and blocks male rats, naloxone Importantly, castrated castrated that in in evidence thresholds stimuli intrahippocampal also nociceptive thermal is increases that to There given response 2004). mediated, of in 3 al., centrally olds effects or et DHT, are the testosterone, (Aloisi thresholds that of dependent nociceptive administration suggesting sex in females, not stimuli increases & are in mechanical androgen-induced (Borzan processes licking to testosterone nociceptive formalin-induced with response on reduces treated in androgens also rats thresholds Testosterone castrated nociceptive 2006). to Mulgaonker, lower Fuchs, relative & have Pednekar inflammation 2006; rats carrageenan-induced Fuchs, male following & electrical (Borzan castrated and replacement thermal Moreover, testosterone to response given 1994). nociceptive in rats in thresholds involved castrated nociceptive are lower than have activity stimuli adults androgenic as and but castrated testosterone rats 2006), Male that processes. al., evidence ample et yield are Daniell populations. studies There 2011; Preclinical heterogenous 2009). al., and sizes Zylicz, et sample Studies & Aloisi small Animal Roantree 2015; by 3.2.2. 2017; al., to limited al., et relative is Basaria et literature therapy (e.g., (Raheem that replacement of findings population testosterone much these control of to a initiation typically exceptions to the hypogonadism several and upon with men baseline analgesics instance, own For opioid opioid use. their of for of intake frequency need or their the dose decreases reduce in generally reductions treatment for testosterone allowing sensitivity, α- nrsaeil( H eaoie nrae oietv thresh- nociceptive increases metabolite) DHT (a androstanediol 8 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. ay n h eritreit oeo h iutr Hn ta. 92 ergi ta. 92.Confirming 1982). al., et Petraglia 1982; al., et (Hong pituitary pitui- the anterior pituitary, of In whole lobe examined. plasma, neurointermediate region in the beta-endorphin the beta-endorphin and increases tary, decreases on consistently castration activity depend androgenic instance, pituitary, concentrations For and concentrations. beta-endorphin plasma in on particularly androgens regions, many of literature. effects existing qualitative the from The evident effects are These patterns 2008). Wood, general Beta-endorphin 2015; some al., 3.4.1.1 peptides but et opioid Mhillaj loci, of 2012; neuroanatomical Hallberg, concentrations across & the vary Nyberg influence see often review, androgens (for exogenous receptors opioid of and administration and castration Both Peptides Opioid 3.4.1 Receptors and Peptides 2019). dopamine Opioid al., (Vel´asquez morphine-induced 3.4 et adulthood rats receptors, testos- in opioid female of preference mu or dose place of male conditioned bolus expression either morphine-induced responses single the in or opioid-mediated a alter effects, on that not locomotor given androgens does morphine-induced effects, of 1 the release, developmental day effects antagonize than postnatal the functionally rather on can of activational terone androgens Some to that Chronic conditions. attributed suggesting 2000). some be 2000), al., under can al., et opioids et Harlan mu (Harlan 2001; of putamen al, effects caudate et the increase morphine-induced D’Souza acutely in decreases (e.g., agonists on significantly regions opioid androgens brain mu exogenous multiple rats, with in male treatment expression intact gene In JunB 2008). al., and et Endoge- (Ahanagar antinociception. treatment opioid-induced testosterone in differences GIRK sex in for decreases of GIRK responsible effector increase partly post-synaptic androgens least primary at nous the are without are 2006). and (GIRKs) or al., opioids channels (with mu et potassium castration (Craft rectifying and inwardly activity 2014), protein-coupled locomotor Wood, G morphine-induced & activity alter Cooper locomotor not 2003; (C´el´erier does morphine-induced al., mice replacement) alter et and testosterone not rats mice do male male both androgens intact Moreover, intact in exogenous in 2004). Woods, hypothermia contrast, morphine-induced & In increases (Peters these nandrolone 2003). naltrexone and (C´el´erier exposure of antagonist, hamsters, al., administration receptor Repeated male et chronic opioid AAS 2006). in and the from activity Wood, acute by lethality locomotor 2004; blocked and and Woods, be 2004), temperature, can & Woods, effects body (Peters & respiration, heroin (Peters decreases and weeks testosterone two morphine to as of resulting little that as mortality resembles in overdoses significant depression produces autonomic examined. testosterone severe endpoints intracerebroventricular from disparate effects. of the opioid-mediated across self-administration unconditioned consistency 2015b; Chronic influence little al., been report androgens et has Bawor research studies exogenous less available 2019; comparatively and few al., contrast, endogenous The In et 2019). AminiLari both long-term Ho, by 1999; on in reviews al., hypogonadism conducted et (see relevant Yilmaz agonists 2016; clinically Wosnitzer, mu to & recreational O’Rourke leading and males, medicinal identify in of activity 2011). Opioids users to androgenic al., of failing decreases et Effects Peckham use studies Unconditioned 1986; Opioid Indeed, on al., Androgens et identify 2002). to Kepler of al., fail Terner 1996; Effects often al., 2005; et and 3.3. et common 2003, administration Krzanoskwa less Cicero al., testosterone much 2002b; (e.g., are et neonatal mechanism sensitivity al., Stoffel via opioid alternative in 2015; females et an differences of al., sex Cicero masculinization et of mechanisms 2005; by (Bai neonatal androgenic or al., by adult males, eliminated or et of completely 2002) (Cataldo castration or al., 2002) partially et al., are Krzanoskwa antinociception. sex females et 2002b; opioid-induced and linking al., of males studies et between modulation within (Cicero differences androgenic correspondence sex strong to instance, a antinociception For is opioid-induced there in findings, differences contradictory seemingly these Despite 2 xrsinfloigcsrto;hwvr hs ffcscno ervre yexogenous by reversed be cannot effects these however, castration; following expression 2 eeepeso ntebanadsia odi aert seiecdby evidenced as rats male in cord spinal and brain the in expression gene 9 c-fos n uBgn expressi- gene JunB and c-fos Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. es o xml,crncnnrln diitainsgicnl erae C ocnrtosi regions andro- in by concentrations control DCE regulatory the decreases under of significantly are cleavage administration concentrations nandrolone via DCE chronic and example, 1992), into modulations For al., gens. androgen-induced et by transforms (Silberring part, DCE peptide in dynorphin (DCE). explained, enzyme be dynorphin-converting may 2008). in dynorphin al., in et Triemstra Birgner modulations (cf., 2009; Androgen-induced mixed al., is theoreti- et possibility should Silva this dynorphin 2007; for in al., evidence nucleus however, reductions moti- et concentrations; the in androgen-induced dopamine in accumbens involved accumbal and activity nucleus structure increase 2013), dopaminergic cally critical The Carlezon, inhibits a 2000b). & Dynorphin is (Muschamp addiction. al., and and accumbens et dynorphin area reinforcement, tegmental (Johansson decreases drug ventral significantly rats behavior, the vated nandrolone intact from with in afferents treatment accumbens concen- dopaminergic dynorphin receives Chronic nucleus in regions. the increase brain in androgen-induced chronic concentrations other the by to in reversed exception seen fully an trations is and accumbens 1986) nucleus 1989). al., the al., Interestingly, et et reversed the (Fullerton (Molineaux partially DHT in be testosterone can with concentrations decreases with treatment dynorphin these in (7-day) treatment and decreases PAG 1989), (2-day) al., castration and subchronic et Similarly, Fullerton striatum, by 1986; 2000a). al., hypothalamus, al., et (Molineaux the et with pituitary anterior treatment (Johansson in chronic rats concentrations instance, male For dynorphin regions. intact brain increases most significantly in dynorphin nandrolone of concentrations increase males 1983). Androgens intact Hong, in & (Yoshikawa 1983). testosterone pituitary with Hong, anterior treatment Dynorphin & the (Yoshikawa chronic 3.4.1.3. 1983). in that castration concentrations Hong, observation following leu-enkephalin the & pituitary and Yoshikawa is anterior met- findings 1982; decreases the greater these al, in concentrations have to increases et leu-enkephalin exception castration also testosterone) and One (Hong and not rats (but met- rats, females male DHT, both female in metabolite, Intact intact of testosterone observed conditions 2000b). than the those of all) pituitary 2000a; administration not anterior to Chronic al., (but the concentrations et some in these under Johansson leu-enkephalin reduces and and (cf., concentra- 2000a) met- area met-enkephalin al., of gray increases et concentrations nandrolone (Johansson periaqueductal with striatum the treatment and many in chronic hypothalamus in peptides the rats, enkephalin in male of tions intact concentrations increase In androgens regions. beta-endorphin, brain on effects their to Similar beta-endorphin of concentrations increase Enkephalins androgens 3.4.1.2. that tissue. rule and latter plasma general these 1994). collectively, both the taken al., When in to ef- 1986). et (Wardlaw, exceptions this testosterone (Menard and represent with hypothalamus, nucleus treatment findings basal chronic te- arcuate by medial reversed the and rostral is in decanoate, fect concentrations the nandrolone beta-endorphin within increases instance, undecylenate, castration For beta-endorphin Moreover, boldenone conditions. decreases androgens, limited, cypionate, exogenous albeit stosterone the some, with under and treatment beta-endorphin reinforcement in chronic of opioid Finally, in concentrations 2000). implicated decrease al., heavily Androgens region et 2015). a (Harlan Margolis, 1997), in rats & al., concentrations et (Fields male beta-endorphin (Johansson addiction increases intact area significantly in tegmental nandrolone ventral with thalamus the treatment of chronic rats, midline male the intact boldeno- 17b-cypionate in AAS, testosterone the beta-endorphin and with not decanoate, increases also treatment but nandrolone rats androgen 1-de-hydro-17a-methyltestosterone, male manner, chronic intact undecylenate, dose-dependent to instance, ne a androgens For exogenous in concentrations. of 2009). testosterone beta-endorphin administration al., with Chronic increases et treatment DHT. (Pluchino chronic metabolite, testosterone rats by plasma, the (Pe- intact reversed in by testosterone to are concentrations relative with effects beta-endorphin hypothalamus treatment latter lower and chronic The exhibit lobe, following rats neurointermediate reversed gonadectomized pituitary, Similarly, partially anterior 1982). are al., effects et these traglia androgens, for role a 10 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. eetr n 2 nrgncdcessi eetrdniyaeosre ntepeec fa nrgnre- androgen an of androgen presence express the not in does observed that are line re- density cell observation delta receptor a the expressing in in by observed decreases naturally evidenced are androgenic as lines density (2) receptors receptor cell and androgen in hybrid receptors decreases of in androgenic independent (1) expression are that delta mRNA effects decreases these receptor treatment Interestingly, Nandrolone delta ceptors. cells. and neuronal in binding 2017). expression al., receptor receptor et opioid (Lee delta rats decrease Androgens male intact dose-dependently in prevents and agonist Receptors flutamide, opioid ganglia Delta mu antagonist, trigeminal 3.4.2.2. a the receptor of in (Zhang androgen effects testosterone mRNA antihyperalgesic the with the receptor with treatment blocks mu treatment chronic of following subchronic upregulation restored Similarly, inflammation-induced is ganglion conditions. 2014). upregulation sensory limited) al., the (albeit and in et mRNA some nerve, receptors under receptor trigeminal mu density of mu the receptor upregulation of opioid on inflammation-induced mu prevents androgens increase castration of in androgens instance, that For effects beta-endorphin noted direct of be the must possible concentrations as It out increased such rule by synthesis. & areas not part, protein Smith in does or in 1986; expression density this explained, al., however, receptor be et regions; those mu Hnatowich can these to in 1992; hypothalamus similar by al., decreases and density part, et midbrain androgenic-induced receptor in (Bergasa Consequently, opioid explained, administration in 2003). concentrations be Yancey, decreases agonist the may chronic compensatory in areas following produce Increases these observed peptides et receptors). concentrations. of opioid mu Piva beta-endorphin some hypothalamic endogenous see endogenous on in of but androgens in density 1991; of al., increases receptor effects et null mu androgen-induced (Limonta of in examples rats decreases for female Androgen-induced 1990 of developmental al., hypothalamus slows et Takayma the recep- administration and mu testosterone in in 1987 increases neonatal density manipulations al., and Castration receptor proandrogenic rats, 1991). mu male either al., in of by et increases hypothalamus (Limonta eliminated the males are in in density differences manipulations tor sex antiandrogenic these by or and females rats, female intact than as n iia ffcsaeosre ntednaegrso atae aspeaal xoe omorphine to exposed prenatally castrated rats vehicle-treated castrated to of relative gyrus hippocampus dentate the the of in regions observed CA3 are ( and effects CA1 concentra- similar lower the and have in rats, rats receptors castrated mu (Takaya- testosterone-treated density, testosterone of receptor with tions opioid treatment mu chronic in by increases blocked decreases Castration see is androgenic midbrain. but effect 1990, and this al., thalamus and et areas, the ma Androgens by these in 2008). blocked in concentrations density fully Spampinato, protein receptor is & receptor mu effect (Guarino opioid this antagonist mu and receptor neuroblas- decrease treatment, human androgenic also In nandrolone an regions. following with brain decreases treatment multiple mRNA simultaneous across receptor cells mu neuronal cells, in toma receptors mu and decrease peptide Androgens In both region. on brain greater effects Receptors androgenic and with Mu compare subtype ligands 3.4.2.1. to across using data inferences. sufficient differ functional studies are draw that Later there to receptors cases, 1985). concentrations all opioid Fishman, testosterone not on with but & treatment effects some by Han reversed androgenic is 1979; reported effect this &Fishman, specificity and Han rats, male (TP; intact to propionate [ relative ligand, brain whole opioid in nonselective receptors the using studies Early exhibits treatment. Receptors which androgen Opioid si- 2007), following 3.4.2. treatment al., concentrations et nandrolone dynorphin (Magnusson contrast, in accumbens In the decrease nucleus significant 2007). including the a al., treatment, in concentrations et androgen DCE (Magnusson following increases PAG concentrations gnificantly and dynorphin hypothalamus in putamen, increase caudate an exhibit typically that lmeo´ ta. 03.Fnly natml asehbtlwrm eetrdniyi h hypothalamus the in density receptor mu lower exhibit rats male intact Finally, 2003). al., Slamberov´a et ˇ lmeo´ ta. 02fropsn ffcsi ueircliuu) ute supporting Further colliculus). superior in effects opposing for 2002 Slamberov´a al., et ˇ 3 11 ]ateoe eotdta atainicessopioid increases castration that reported H]naltrexone, Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. e r oelkl ouehri n te lii pod,weeswmnaemr ieyt s pres- use to likely more are women whereas opioids, illicit other and heroin use to likely more are Disorders Men Use analgesia. by Opioid opioid augmented enhance for be and Implications could sensitivity dose treatment 4.2. measuring pain subsequent opioid diminished tests decrease and endogenous detected, simultaneously determinations and blood is of to dose hypogonadism simple disorders treatment levels initial where by androgen pain guide low cases diagnosed could chronic in Regardless, levels be Moreover, both Testosterone 2018). quickly adjustments. testosterone. to Nieschlag, can total contributor & Hypogonadism and Vorona free analgesics. unrecognized (Christou 2011; opioid an hypogonadism to Hallak, and be sensitivity & fertility may for Souza with activity women, clinically de problems androgenic In 2017; Although to women. androgens leading al., receptors. and exogenous activity, men, men et androgen in androgenic both whereas of effects, for endogenous activation anti-feminizing drawbacks suppress and indirect limiting masculinizing clinically produce or androgens have instance, direct would opioid of by practice effectiveness this increased the intriguing, that blockade be is androgenic analgesia could opioid by enhancement analgesics minimized androgen-induced males. of be in implication can activity obvious females. androgenic One differences in greater sex stimulation to that androgenic (at underappreciated part, additional suggest or but in doses sizeable Some due, males data lower A are in 2.2.1). clinical need analgesia. differences typically and of (Section sex levels and preclinical these disorders comparable that analgesics Indeed, obtain pain suggests opioid to evidence from weight) to of body suffer sensitive body of more to function are a likely as men more are least that women are indicates that and indicate also strongly pain evidence studies to epidemiological sensitive and areas. clinical, more these preclinical, in from making evidence The Converging decision Analgesia practice. future amplify health Opioid guide to public might for androgens or that Implications of clinical possibilities ability 4.1. in several The considered to disorder. sufficiently in point use apparent been above opioid are not reviewed for differences has data risk sex differences the significant these above, and minimize) reviewed analgesia (or number As opioid a health. has to behaviors public sensitivity opioid-mediated and both in personal differences for sex implications to contribute of populations. may androgens human that in observation Implications drug AAS The accumbens Health of and effects nucleus Public behavior abuse-related and the motivated and Clinical in positive-reinforcing in 4. signaling the involved for opioid modulates critically part, kappa negatively is in in account, dynorphin which could decreases 3.4.1), accumbens, androgen-induced (Section nucleus Consequently, above the addiction. noted in receptor opioid As release kappa and compensatory is treatment. dopamine concentrations exception as androgenic dynorphin notable both part, these following in One of in decreases density concentrations. most exhibits explained, In dynorphin which slices). be endogenous accumbens, brain nucleus can in hypothalamic the increases density on castration androgen-induced receptor et of to kappa effects (Magnusson responses in the endopiriform for decreases dorsal brain 2015 and androgen-induced al., most putamen et regions, nandrolone Ruka caudate in chronic see the contrast, also receptors In in 2009, 2009). density al., opioid al., receptor et kappa kappa (Magnusson terminalis increases decreases amygdaloid stria treatment central and hypothalamus, nandrolone shell, pallidus, accumbens globus with nucleus investigation. lateral the single nucleus, treatment in a observed from chronic decreases derived significant is rats, with receptors kappa regions, on male androgens of intact effects the In about known is what in of Much increases androgenic by Receptors induced in Kappa down-regulation decreases 3.4.2.3. receptor androgen-induced by admi- receptors, part, by mu concentrations. in in completely to enkephalin reversed explained, density Similar endogenous is be receptor 1990). effect may opioid al., this receptors delta et and delta (Takayama increases hypothalamus, testosterone castration and is of instance, density midbrain, nistration for receptor thalamus, opioid rats, bulb, delta olfactory In in the regions. decreases brain Androgen-induced across 2000). al., widespread et (Pasquariello antagonist ceptor 12 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. nbt ae n eae odtrietecnrbto fadoest e ieecsi pod reward opioids in differences sex to androgens of contribution and the conducted physiological determine be to both to need females including start will and devel- to reinforcement, studies and males positioned for Preclinical opioid AAS both perfectly risk abuse. between in is in the AAS link androgens research of on clear Preclinical characteristic of androgens a concentrations absent. role of supraphysiological indicate is effects the relationship data equivocal, reinforcement, the examining causal epidemiological are regarding a and the systematically data of conclusions reward Although preclinical evidence draw opioid available abuse, disorder. to on opioid The use difficult androgens substance is literature. a of it laboratory oping time effects human this the and at regarding clinical and information the are of problematic in opioids minimizing dearth particularly thus if a reduced, Even liability. is be abuse could analgesia. There andro- and opioid opioid indeed, tolerance of for pain as analgesics; dose need such androgen-augmented the opioid effects the that with of side suggest negate augmentation trials data may androgenic clinical available to analgesics the begin necessary, limited nonopioid hypogonadism. pa- to be of borderline data dosing not augmentation or sufficient optimal need genic undiagnosed now trials and with Such are tolerance, men opioid there in pain therapies. increase management populations, levels, and pain these testosterone sensitivity maximize pain In of to decrease evaluation needed androgens Clinical are that rameters suggesting 3.2). literature a as of (Section cycle body analgesia estrous sizable the a same include model these is must on There working differences hormones a sex ovarian in explore create of differences explicitly role to sex that complex to variable. the data to studies the biological that degree Future enough androgens by of suggest complicated not females. the contribution further strongly in are The and is proteins data there observations. receptors available stage, but empirical and the developmental of peptides specific, activational All number opioid or region or limited and age specific. the developmental age explain region across predominately to are are differ are androgens androgens 2.4 androgens androgens of is of (Sections of effects of There effects proteins effects central effects these and research. the the in the peptides further whether which differences whether opioid for unclear sex nature, endogenous is ripe reported in It to is both contribute that of 3.4). androgens area concentrations and that central an and is modulate receptors, behavior androgens opioid opioid-mediated that on evidence androgens ample for of candidates role them avoided. The making be 2010), must al., effects Directions et feminizing Future Mella or 5. 2016; masculinizing al., potential Finasteride where et male- testosterone. trials Hirshburg (i.e., clinical of 2014; future levels alopecia Charrette, normal androgenic & with (Gupta and men tioning prostate) impact 5 5 enlarged exclusively a other or (i.e., is and primarily to hyperplasia finasteride sensitive that prostatic is example, baldness) seeking For treat pattern opioid feasible. to or because intake be used men- problematic opioid might inhibitor be if As interventions problematic also Regardless, to some disorders. would men. lead manipulations, antagonists use cisgender would receptor in androgenic opioid receptors Androgen effects of individuals. androgen feminizing treatment of produce at determined majority they the agonists be the indirect in to in or remains aid effects direct It side to of opioids. manipulated are a and administration who is be AAS above, males, opioids both tioned can adult of misuse activity young simultaneously misuse interventions and androgenic and the early adolescent AAS whether that an utilize be similarly, misuse would should suggesting and to interventions programs likely opioids, abuse, these prevention mostly opioid of illicit drug-abuse for target 3.1.1), and obvious factor (Section An licit risk abuse appropriately. notable a AAS both nonetheless is for drugs. of is AAS factor of It abuse of risk classes misuse androgens. the pharmacological the of two with that role the correlated Given between the highly relationship at unknown is looking in there still AAS data and hormones but examined, clinical of gonadal underappreciated extensively and abuse of been preclinical not role the has both many The disorders that of to use role. paucity opioid due prominent and a likely are most use is observations more opioid the much in These playing differences are use. likely sex women drug mediating factors Moreover, their psychosocial 2.1.1). from with (Section consequences disorder factors, psychosocial use negative opioid experience an to develop and opioids α- euts niiosd o rdc eiiigeet n aelmtdeet nsxa func- sexual on effects limited have and effects feminizing produce not do inhibitors reductase 13 α- reductase Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. hrp n podtprn o podidcdhpgnds mn ainswt hoi ocne pain: noncancer replacement chronic Hormone with (2019). patients W. among J. hypogonadism review. Busse, opioid-induced systematic & affects for L., a Testosterone Wang, tapering R., (2004). opioid Couban, (2011). and C. S., therapy G. Massafra, Craigie, P., & Pari, Manjoo, rats. M., & M., female A. AminiLari, and Padova, male De . in . P., . differently Fiorenzani, responses S., I., formalin-induced Ceccarelli, Mameli, M., G., patients. A. Sindaco, pain Aloisi, chronic A., male Suman, hypogonadic Endocrinology M., and morphine-induced Biology Carlucci, M. in therapy I., Meriggiola, men. replacement & Ceccarelli, and Hormone A., women M., Bertaccini, transsexual A. in I., Aloisi, pain Ceccarelli, changes R., administration Stefani, hormone A., in S60-S67. Cross-sex Costantino, properties V., affective (2007). rewarding Bachiocco, C. motivation. has sexual M., Testosterone of A. basis (1994). Aloisi, morphine biological M. on the Hines, gender for & implications and G., housing rats: M. of male Packard, effect M., The G. (1978). Alexander, F. P. Hadaway, & rats. B., in R. self-administration (2004). Coambs, gene E. K., blockade. J. GIRK2 B. Grant, opioid Alexander, & on to C., responses hormones Kirschbaum, W., hypothalamic-pituitary-adrenocortical steroid S. and medicine Kim, gonadal matic G., pain of Nordehn, in D., differences Effects Ellestad, Sex E., L. (2008). Wittmers, M., M. system. al’Absi, nervous Jorjani, central rat & the B., in transcription Kazemi, N., Ahangar, References Interest: of the Conflict approved authors All draft. for manuscript. final this manuscript the writing the on submit Acknowledgements: collaborated to MS decision and JLS the manuscript. drafts. in final initial or the manuscript wrote and the of writing Contributors: the in role no publication. had NIH The Source: Funding of Role under- public remain and that translational Disclosures meaningful topics and carry several Funding may identifies that research review of This lines management several impact. disorders. clinical proposes health and use better A area to substance this lead and reinforcement. in would studied pain and outcomes reward chronic on these opioid-mediated pain both influence consequences to to androgens of functional regard how sensitivity in of andro- to and particularly understanding circulating lead analgesia, relevance, greater clinical opioid of receptors have to control and consequences sensitivity tonic these peptides sensitivity, of under these Some are of behavior. receptors opioid-mediated modulations and Androgen-induced peptides opioid gens. disorder. of as use serves concentrations opioid activity an Endogenous androgenic developing estrous of whether likelihood determine the Conclusions the to and in needed 6. hormones influence) be ovarian protective ultimately (or of will factor influence studies risk potential of a the types These consideration into cycle. taking reinforcement, and , A n Pdvlpdteie.T n L efre h eiwo h rmr literature primary the of review the performed JLS and TP idea. the developed TP and MAS 66 2,198-206. (2), anMedicine Pain h uhr iht hn r alShitfrcmet napeiu eso of version previous a on comments for Schmidt Karl Dr. thank to wish authors The ocnitdeclared. conflict No Psychopharmacology , hswr a upre yNHGat A434 A375 n DA045714. and DA031725, DA045364, Grants NIH by supported was work This 9 1,1-10. (1), , 20 2,301-313. (2), ersineletters Neuroscience , 58 14 2,175-179. (2), ersineLetters Neuroscience eairlneuroscience Behavioral , 431 3,201-205. (3), , 361 13,262-264. (1-3), , 108 Pain Reproductive Psychoso- 2,424. (2), , 132 , Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. ekr .B,&Ko,G .(06.Sxdffrne naia oes ou naddiction. on focus models: animal addic- in and differences Sex reward mediating (2016). F. mechanisms G. neural Koob, reviews in & differences B., J. Sex Becker, of development (2019). Perinatal E. (1979). Chartoff, R. & (2015a). tion. Guillemin, B., D. & brain. J. Desai, A., rat Becker, & Mauss, the E., in receiving F. systems . users . enkephalin-containing Bloom, . opioid endorphin-and J., C., the among W. Plater, Shoemaker, functioning C., A., social D. Bayon, and Marsh, study. health, cohort J., multicenter use, Daiter, a substance M., treatment: Varenbut, in B., differences B. Sex Dennis, M., Bawor, (2015). L. J. Rhudy, & L., J. DelVentura, women. L., healthy E. in Terry, and hypoalgesia L., Sex with K. (2002). associated Kerr, J. is L., sex M. pain testosterone B. Picker, of in Kuhn, & variation Importance S., C., Natural Syvanthong, Palit, L., (2001). J., E. E. J. Roach, Bartley, M., M. J. to Picker, Terner, sensitivity determine D., & strain C. M., rat Cook, R. C., A. Craft, Barrett, M., J. receptor W. opioids. mu-opioid Terner, of the Ling, peripheral effects D., at in antinociceptive & differences efficacy C. Sex relative Cook, (2015). . and . X. C., . He, model. A. L., & pain B., Barrett, Haynes, persistent Li, orofacial Z., L., multisite rat Lu, in national Stroud, Y., analgesia a Li, E., mediated from X., R. results Zhang, X., Carter, dependence: Bai, opioid H., with A. women and Wahlquist, men trial. L., effectiveness of Findings profiles R. Comparative opioids: Payne, prescription (2011). and E., Gender Health. S. and (2010). Use T. Back, K. Drug Brady, on & Survey N., National A. require- Simpson, the L., morphine from R. in Payne, E., differences S. age-related Back, Sex-and of (2005). effects B. relief. rewarding Riou, pain Similar 156-160. & postoperative P., for Coriat, (2002). individuals. ments latency N., O. attack Salvi, Pellicer, long F., and & Aubrun, short as Mart´ınez-Sanchis, S., of rated A., mice properties Salvador, in Rewarding testosterone T., (2000). study. M. E. pilot Gonzalez-Bono, Arnedo, a & mice: S., male Martinez-Sanchis, intact in A., in hostility testosterone Salvador, of T., degrees various M. Becoming and Arnedo, relation- levels The 2. Mor Trpv1, (2018). careers. testosterone, M. males. free addict Chakhnashvili, healthy sensation, & in young pain D., differences Mzhavanadze, thermal Sex between M., ship Tsagareli, (1987). I., H. Kvachadze, M., W. Apkhazava, McGlothlin, & L., Y. Hser, addicted. D., M. Anglin, ao,M,Bm,H,Dni,B . ltr . ose,A,Vrnu,M,...&Coe .(2015b). M. Coote, & . . . M., Varenbut, A., Worster, C., meta-analysis. Plater, and review B., systematic a , B. (2015). users: Dennis, opioid O. in M. H., suppression Testosterone Martel, Bami, & M., controlled . . randomized Bawor, . a C., deficiency: Cahalan, androgen K., opioid-induced Teeter, with E., men P. trial. in Knapp, replacement D., testosterone of Alford, Effects G., T. Travison, S., Basaria, 149 , Neuropsychopharmacology Pain 1-9. , 31 , 68 h mrcnjunlo rgadachlabuse alcohol and drug of journal American The 8,730-739. (8), , 156 2,242-263. (2), 2,280. (2), h mrcnjunlo rgadachlabuse alcohol and drug of journal American The erinMdNews Med Georgian μ podrcpo ntedvlpeto oeac n rs-oeac othe to cross-tolerance and tolerance of development the in receptor opioid , Psychopharmacology 44 κ podidcdantinociception. opioid-induced 1,166-183. (1), h ora fteAeia oit fAnesthesiologists of Society American the of Journal The ilg fsxdifferences sex of Biology , 283 hraooyBohmsr n Behavior and Biochemistry Pharmacology 109-14. , ditv behaviors Addictive 15 , 158 lSone PloS , 13 2,154-164. (2), 12,59-71. (1-2), , 6 ri Research Brain Psychopharmacology , , 1,21. (1), 37 10 dito biology Addiction , 5,313-323. (5), 3,e0122924. (3), 35 1) 1001-1007. (11), rgadachldependence alcohol and Drug , h lncljunlof journal Clinical The 179 , 1,93-101. (1), , , 160 Pharmacological 65 7 4,373-379. (4), 2,327-332. (2), 2,170-181. (2), , 103 (1), Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. ffcso nblcadoei triso h erdciesse fahee n erainlues a users: recreational (2017). S. and Tigas, athletes pain & of and G., system Gender Mastorakos, reproductive A., (2002). the Tsatsoulis, N. on P. G., steroids Wang, Markozannes, androgenic & A., a anabolic P., P. of L. analgesia: Christou, Effects patient-controllediv Ger, A., postoperative K., M. for Liu, Christou, patients. requirements C., Chinese the 2,298 C. of with Hung, survey associated prospective H., are L. movement Chow, upon potential Y., their ef- and Y. Side function receptor (2003). Chia, opioid kappa L. in B. differences Sex addiction. Strom, on (2015). & M. impact Mavrikaki, race. B., & and H., D. E. gender, Carr, Chartoff, age, R., of Effect Boston, men than M., administration: short-term morphine Baumgarten, Therapeutics during more T., opioids require J. of and Farrar, fects pain S., more M. experience analgesia. Women Cepeda, of (2003). degree B. similar D. a achieve Carr, to & S., M. mice. Cepeda, in nan- dependence of and Effects tolerance (2003). responses, R. , morphine Maldonado, acute & on F., drolone Nyberg, S., Casta˜n´e, Ghozland, T., C´el´erier, A., M. Yazdi, E., the of phases and hyperalge- differences and Sex inflammation (2000). peripheral to A. neurons M. dynorphin Ruda, cord & spinal of K., response sia. Malsnee, carrageenan- the Q., on alter Ling, cycle testosterone estrous J., of Miller, effects H., activational Bradshaw, and analgesia. Organizational morphine and (2006). pain N. inflammatory P. induced withdrawal: Fuchs, and hormones. & tolerance gonadal J., hyperalgesia, of analgesia, Borzan, roles opioid in and differences action Sex of (2010). mechanisms B. central Testosterone Kest, & (TRiUS). (2012). J., States M. R. United M. Bodnar, the Miner, in & Registry H., Testim the Kushner, users: D., Medicine opioid Nguyen, among K., outcomes R. therapy administration Bhattacharya, replacement nandrolone M., sub-chronic Khera, to G., sex response Blick, in underlies challenge. shell activation amphetamine accumbens receptor subsequent nucleus a opioid rat and the mu PAG in HVA and (2007). DOPAC M. Kindlundh-H R. C., Craft, Birgner, & antinociception. M., morphine mu-opioid M. Central in Morgan, (1992). differences A. cholestasis. A., E. of Jones, S. & model Bernal, G., rat M. a Swain, H., in Xu, down-regulated J., are Vergalla, B., receptors R. Rothman, V., N. Bergasa, aad,G,Bra,S,Mroiz . gw,S,Rgat,A,Pa,D . onr .J (2005). J. R. Bodnar, & of W., effects rats: D. female Pfaff, in A., replacement. analgesia morphine estradiol Ragnauth, systemic and S., ovariectomy and Ogawa, hormones adult cocaine sex gonadal Intravenous A., of and (2002). Markowitz, manipulation phenotype K. S., Organizational intake: N. Bernal, saccharin Dess, G., differential & Cataldo, C., for U. bred Campbell, selectively J., rats W. in differences. and Lynch, self-administration processes D., heroin Nonassociative A. (1996). and Morgan, L. E., depen- M. M. Boechler, steroid Carroll, & Anabolic-androgenic C., testosterone. G. (1989). by Abrahamsen, conditioned C. S., preferences Fuelling, H. place Stock, & J., P., B. T. Caldarone, Beresford, C., F. Blow, dence. J., K. Brower, 465 Pain 12,69-81. (1-2), h ora fciia psychiatry clinical of Journal The , , 85 13 Psychopharmacology , 12,93-99. (1-2), 5,688-698. (5), 74 2,102-112. (2), rnir nneuroscience in Frontiers geg .M,Nbr,F,&Bergstr & F., Nyberg, M., A. ¨ ogberg, , 161 3,304-313. (3), nshsa&Analgesia & Anesthesia eaiua ri research brain Behavioural . ersineletters Neuroscience 50 aainJunlo Anesthesia of Journal Canadian ri research Brain h scooia Record Psychological The 1,31–33. (1), , 9 466. , 16 Neuroscience ora fhepatology of Journal , omnsadbehavior and Hormones 1059 m .(07.Atrdetaellrlvl of levels extracellular Altered (2007). L. ¨ om, , , 412 97 , , 143 1,13-19. (1), 177 2,168-172. (2), 5,1464-1468. (5), , 3,885-893. (3), uoenjunlo pharmacology of journal European 1,126-133. (1), 46 2,373. (2), , 49 , lnclPamclg & Pharmacology Clinical 15 3,249. (3), , 12,220-224. (1-2), 58 1,72-81. (1), Pain Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. rf,R . lbri .M,&Ru,D .(00.Kpaoii-nue irssi eaev.male MK-801: and vs. morphine of female effects dimorphic in Sexually diuresis in (2002). opioid-induced M. differences M. mechanisms. Kappa Garcia, Sex and-independent & steroid-dependent (2000). E., sex (1999). R. J. Harlan, J. N., D. S. D. Raub, D’Souza, King, & & M., I., C. T. Ulibarri, rats. Walpole, M., rat. E., the R. R. in Craft, dependence Bartok, and A., tolerance determines J. morphine gender, of not Stratmann, development but frequency, M., Reinforcement R. (1998a). morphine. differences Craft, A. of Sex S. effects Bernal, stimulus (1998b). & discriminative M. L., to J. C. sensitivity Morgan, Hanesworth, M., & R. W., Craft, the J. of Harding, effects S., Behavior diuretic effects and J. stimulus and Biochemistry Boyer, discriminative stimulus J., in discriminative P. differences Kruzich, in Sex M., (1996). R. A. Craft, stimulus J. rat. discriminative Stratmann, the on & in gonadectomy of W., morphine of P. effects of Effect Kalivas, locomotor M., (1999). in rats. R. E. male differences Craft, R. versus Sex female Bartok, in & (2006). morphine M., K. of L. M. effects Heideman, Pinckney, M., & R. antinociception: P., Craft, S. opioid Hart, in rat. L., differences the J. in Sex Clark, morphine M., opi- (2001). R. of A. Craft, effects S. motoric Bernal, and morphine & discriminative, agonists. with M., reinforcing, interaction R. analgesic, testosterone in Craft, differences : Sex and Androgens (2008). oids. M. (2014). R. I. of Craft, elevation R. rats. opioid-induced Wood, male in & in self-administration differences E., sex-related S. determine Cooper, aries). hormones (Ovis Steroidal sheep in in threshold (1998). differences nociceptive Sex-related J. in (2000). efficacy C. differences J. and intensity, M. Cook, sex stimulus Picker, nociceptive & in genotype, R., rat steroids J. of Bowman, the of importance L., at opioids: Role E. of Roach, effects C., antinociceptive (2002b). A. the Barrett, R. D., E. C. Cook, Meyer, physical of & effects. expression L., organizational the Therapeutics O’Connor, and mental in activational B., differences analgesia: Gender-linked Nock, morphine-induced J., (2002a). T. R. E. Cicero, Meyer, rat. & properties the B., antinociceptive in the Nock, dependence in J., differences T. Gender-related Cicero, (1996). properties R. reinforcing E. the Meyer, in & morphine. differences B., of Gender Nock, (2000). J., R. T. E. Cicero, Meyer, & J., Ogden, morphine. T., of Ennis, self-administration J., intravenous the T. in Cicero, differences Gender (2003). agonists. R. E. Meyer, mu & of C., S. Aylward, J., T. Cicero, 017-0709-z meta-analysis. and review systematic hraooyBohmsr n Behavior and Biochemistry Pharmacology xeietladciia psychopharmacology clinical and Experimental μ podreceptor. opioid rgadachldependence alcohol and Drug ora fPamclg n xeietlTherapeutics Experimental and Pharmacology of Journal Behavior and Biochemistry Pharmacology hraooyBohmsr n Behavior and Biochemistry Pharmacology , 300 hraooyBohmsr n Behavior and Biochemistry Pharmacology hraooyBohmsr n Behavior and Biochemistry Pharmacology eaiua pharmacology Behavioural Psychopharmacology 2,695-701. (2), , 61 Neuroreport 4,395-403. (4), prsMedicine Sports , 63 e eln eeiayjournal veterinary Zealand New 3,215-228. (3), , , 25 rgadachldependence alcohol and Drug 150 , 65 7,521. (7), . 4,430-442. (4), , 17 1,53-59. (1), ora fApidPhysiology Applied of Journal 16 κ , 47 , podaoitU9 9 nterat. the in 593 U69, agonist opioid 5,376. (5), 65 eaiua Pharmacology Behavioural 9,16-83 https://doi.org/10.1007/s40279- 1869-1883. (9), 1,91-96. (1), Psychopharmacology , 85 , , 74 72 4,850-858. (4), ora fPamclg n Experi- and Pharmacology of Journal 3,541-549. (3), 3,691-697. (3), , 279 , , 2,767-773. (2), 46 53 2,68-71. (2), 2,95-109. (2), , , . 92 κ 143 n mxdaction’ ‘mixed and 2,493-503. (2), 1,1-7. (1), Pharmacology Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. ilni,R . ig .D,RbioDsla .C,RhmWlim,B,&RlyII .L 20) Sex, (2009). L. J. III, Riley & findings. B., experimental and Rahim-Williams, clinical C., recent of M. review Ribeiro-Dasilva, a D., pain: C. and reward. gender, King, opioid B., Understanding R. Fillingim, (2015). B. E. Margolis, & 217-225. the L., in differences H. sex Fields, between Dissociation opioid- (2006). T. in D. of differences Lysle, Sex effects , & physiological J., (2003). and A. T. Sparrow, behavioral, D. J., immunological, Lysle, M. Picker, & C., A., J. K. part Elliott, Carrigan, in J., hypersensitivity. due C. contact be Nelson, of may J., enhancement effects M. induced Picker, analgesic C., and J. anti-anxiety Elliott, Testosterone’s 5 (2005). its A. of C. actions Frye, to & L., K. Edinger, 5 Effects Its Cognitive-Enhancing of and Actions Anxiolytic, to Analgesic, , Testosterone’s Part in (2004). C. Due A. E. Be C. Strain, May Frye, & & E., L., G. K. phenotypes. Bigelow, Edinger, withdrawal (2007). A., opioid D. A. Tompkins, meaningful S. R., clinically J. and Aicher, Schroeder, different biology & S., of A. W., evidence Huhn, Preliminary C. M., (2018). Winkler, E. Weerts, M., E., rats. D. K. female male Dunn, and Connor, male in A., of dihydrotestosterone J. medulla ventromedial Harris, and rostral X., Neurology the estrogen in A. self- of receptors Oliveira, opioid testosterone Self-administration De Kappa to T., (2006). sensitivity C. I. Drake, enhance R. androgens Wood, Circulating & hamsters. N., (2004). A. I. syndrome: DiMeo, R. withdrawal Morphine Wood, hamsters. male & (2005). in administration N. N., G. A. mice. Balerio, female DiMeo, & and male C., prepubertal M. in Behavior Rubio, system and the K., dopaminergic in the A. differences of Kemmling, sex-related involvement of L., Lack S. (2001). mice. Diaz, N. in G. syndrome Balerio, withdrawal & morphine C., review. the 75-79. M. comprehensive of Rubio, a baclofen K., by infertility: A. prevention male Kemmling, and L., steroids S. Anabolic Diaz, (2011). Dopaminergic J. Hallak, (2009). and of & P. international administration appetitive L., J. intranasal G. Huston, as by Souza, & Testosterone enhanced de E., accumbens T. (1992). nucleus Buddenberg, E. and B., neostriatum N. Topic, testosterone. in C., Poll, activity Mattern, de serotonergic A., Van of and M. consequences & Silva, and L., Souza effects. risks de J. dose-dependent psychological sex-and Slangen, and rats: E., in Medical stimulus Jansen, discriminative (2012). R., R. Beun, in Chou, De therapy & patch women. testosterone R., in of therapy B. opioid study Stacey, long-term pilot D., Open-label and B. animal (2006). Darnall, A. opiates: deficiency. N. to androgen Mazer, responses opioid-induced & with Sex-specific R., men Lentz, (2008). W., E. H. Sarton, Daniell, & R., A. Waxman, studies. human B., Kest, A., Dahan, 185 118 1,66-75. (1), 1352. (6), , 25 omnsadbehavior and Hormones , 500 1,e12680. (1), uoenNeuropsychopharmacology European , , 108 82 nshsa&Analgesia & Anesthesia 3,465-476. (3), α- 4,601-607. (4), eue eaoie ntehippocampus. the in metabolites reduced 1) 8016.https://doi.org/10.1111/j.1464-410X.2011.10131.x 1860-1865. (11), hraooyBohmsr n Behavior and Biochemistry Pharmacology , 49 anMedicine Pain α- 4,519-526. (4), , eue eaoie nteHippocampus. the in Metabolites Reduced 107 h ora fPain of Journal The 1,83-95. (1), ora fivsiaiedermatology investigative of Journal , 19 , 18 13 κ- 1,53-63. (1), 9,1181-1211. (9), and Psychoneuroendocrinology δ- pod nFshrrats. Fischer in opioids hsooy&behavior & Physiology , 7 h ora fpain of journal The 3,200-210. (3), eaiua Pharmacology Behavioural rnsi neurosciences in Trends , 79 hraooyBiochemistry Pharmacology 2,383-389. (2), eairlneuroscience Behavioral ora fComparative of Journal , Psychopharmacology , 121 , 52 30 , 10 4,629-634. (4), 5,1053-1059. (5), 5,418-430. (5), 5,447-485. (5), Addiction , , 38 12 BJU (1), (4), Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. foii eetrlk eetrmN n ooaiainwt srgnrcpo RAi ern fthe of neurons in mRNA distribution receptor rat. the estrogen the in with differences in Sex-related colocalization caudalis and (2003). nucleus S. mRNA trigeminal S. receptor spinal Mokha, 1 & receptor-like L., opioid S. Petersen, Experimental of P., humans. Shughrue, (2004). A., in R. C. Staud, analgesia Flores, & D., Anesthesiologists D. in of Price, differences M., Society C. American sex Campbell, the no L., reveal T. (2005). but Glover, models R. J., responses Staud, pain T. cardiovascular & Ness, and D., B., effects D. R. Price, side Fillingim, A., in B. differences Hastie, sex M., C. analgesia. pain: Campbell, not experimental L., T. and Glover, responses J., Morphine T. Ness, B., R. Fillingim, refil,S . ros .J,Gro,S . re,C . rp,F,MHg,R . il,G M. G. Miele, & . literature. . . the K., of R. review abuse McHugh, A F., who women: Kropp, Women in A., outcome dependence (2009). C. and alcohol Green, retention, F. and entry, M., treatment S. S. abuse Gordon, Butler, Substance J., (2007). A. & Brooks, H., F., S. S. Greenfield, Budman, A., Version Licari, Index-Multimedia database. Severity G., Addiction opioid M. the from J. Findings opioids: trial. Serrano, prescription placebo-controlled C., and T. An- randomized, Green, & double-blind, G., body P. a improved Andersen, Endocrinology hypogonadism perception: T., of male pain Graven-Nielsen, Journal opioid-induced not E., of Bendix, but therapy L., replacement composition Testosterone L. (2020). Christensen, M. B., dersen, program. H. treatment Vaegter, maintenance D., a Glintborg, to admission on life of Research quality Addiction R health-related G., in Kemmler, differences M., Ertl, der Y., Riemer, M., Sweden. S. Giacomuzzi, in lobe anterior and and steroids steroids anabolic-androgenic Gonadal of Dependence use Alcohol (1989). Dual W. (2010). J. action A. Funder, Rane, of G˚arevik, & & site A., N., rat. a J. male Clements, the as I., in accumbens A. dynorphin nucleus Smith, The J., M. 5 (2002). Fullerton, its B. Svare, and & testosterone R., of Behavior Rosellini, properties E., rewarding M. for Rhodes, A., C. Frye, ars .A,Cag .C,&Dae .T 20) ap podrcposi a pnlcr:sex-linked cord: spinal rat in receptors opioid Kappa (2004). T. Androgenic–anabolic C. Drake, (2000). & M. of C., P. differences. M. role distribution Chang, Garcia, possible A., & J. striatum: D., Harris, rat D’Souza, the S., content. in C. receptor expression Lynch, opiate c-fos research E., brain morphine-induced and H. rat blunt castration Brown, male steroids E., upon affects content R. Castration Harlan, receptor (1985). opiate J. brain Fishman, rat & docrinology in F., Changes E. Hahn, (1979). J. Fishman, dutasteride. replacement. & and testosterone F., 5 finasteride E. of of Hahn, safety assessment and benefit–risk efficacy and The meta-analysis treatment (2014). SH-SY5Y dermatological network A. in a Charrette, expression & alopecia: receptor K., opioid A. mu Gupta, decreases Nandrolone (2008). cells. S. neuroblastoma Spampinato, human & G., Guarino, , , 853 74 , 41 h ora fPain of Journal The 1,119-127. (1), 1,99-104. (1), 1,60-63. (1), rgadachldependence alcohol and Drug , , 11 109 , Neuroscience 2,69-75. (2), , 86 , iceia n ipyia eerhcommunications research biophysical and Biochemical 13,144-146. (1-3), 25 ora fseodbiochemistry steroid of Journal 182 Neuroreport 1,1-21. (1), 2,156-161. (2), 6,539-548. (6), , 6 , 2,116-124. (2), 124 , , 19 100 4,879-890. (4), Neuroscience 1) 1131-1135. (11), , 103 5,1263-1270. (5), α- 19 12,65-73. (1-2), eue metabolites. reduced slr . itrue,H,&Kr,M 20) Gen- (2005). M. Kurz, & H., Hinterhuber, H., ¨ ossler, , , 118 32 2,303-308. (2), 3,769-778. (3), α- euts niiosi androgenetic in inhibitors reductase hraooyBohmsr and Biochemistry Pharmacology nshsooy h ora of Journal The Anesthesiology: , 90 ® 3,819-823. (3), onc prescription Connect β- endorphin. ora of Journal rgand Drug European European Neuroen- Brain Drug Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. oaso,P,Hlbr,M,Knlnh . yeg .(00) h ffc noii etdsi h rat the in decanoate. peptides nandrolone opioid steroid, on androgenic effect anabolic The the (2000a). gonadectomy with F. and bulletin treatment Nyberg, gender, chronic & aging, A., after among Kindlundh, brain, Interactions M., (1993). Hallberg, P., J. Johansson, R. rats. in Bodnar, antinociception & Persons morphine L., for upon M. Outcomes effects Treatment Cooper, in K., Differences A. Sex-Based Islam, (2019). E. K. Disorder. opioid-based Dunn, in Use & differences Opioid S., sex-based With M. of Berry, review S., Systematic A. Huhn, (2018). E. K. of Dunn, administration & Chronic S., effects. M. (2016). in Berry, H. 7 S., L. LVV-hemorphin A. Addiction. Chow, with Huhn, & correlation 3. J., without Y. careers. Chen, dependence addict Y., morphine T. in rats. to Huang, differences susceptibility H., Sex increases Y. Chen, nandrolone (1987). K., W. Y. E. M. abuse Huang, alcohol Booth, and & drug D., [Met5]- of M. pituitary journal rat Anglin, American the I., in Y. difference rats. Sex-related Hser, female and (1982). A. male gonadectomy. C. in by Lamartiniere, tolerance level—altered & enkephalin Morphine K., Yoshikawa, (2004). S., P. J. E. Hong, Wala, and & male Behavior W., in and J. hyperalgesia Biochemistry Sloan, morphine-induced macology R., of J. Characterization Jr, Holtman (2005). P. E. Wala, & rats. female R., J. syndromes. withdrawal Jr, Endocrine Holtman (2003). P. G. Chrousos, of & efficacy K., , Pacak, and E., consequence, Z. prevalence, Hochberg, (OPIAD): deficiency naloxone. androgen [3H] and Opioid-induced reduces morphine replacement. stress testosterone chronic (2019). Cold-restraint and W. K. acute (1986). Ho, B. of G. Influence Glavin, membranes: & brain K., rat research Kiernan, to S., binding F. review. Labella, effects systematic Adverse R., a (2016). M. S. Hnatowich, dutasteride): J. (finasteride, Reichenberg, dermatology & inhibitors C., aesthetic reductase A. and Gavino, 5-alpha clinical A., of C. safety Therrien, A., and P. Kelsey, M., J. Hirshburg, aaaa . rwr .J,Wo,R . usn .I,&Pp r .G 20a.Anabolic–androgenic (2009a). G. H. Jr, Pope & I., J. Hudson, disorder. I., emerging R. an anabolic– Wood, of dependence: Treatment J., steroid K. (2010). Brower, G. G., H. Kanayama, Jr, Pope implications. & its I., and J. hamsters. 6-13. evidence Hudson, (1-3), male Emerging I., in dependence: R. self-administration Wood, steroid J., androgenic testosterone K. Oral Brower, G., Kanayama, (2001). androgenic I. Anabolic R. Wood, (1997). & docrinology F. R., Nyberg, L. & Johnson, K., Karlsson, W., Huang, , affects Q., steroids Zhou, increase androgenic rat. steroids A., Anabolic the Ray, P., in (2000b). Johansson, peptides C. opioid Fahlke, brain & and F., behaviors Behavior Nyberg, defensive S., intake, A. alcohol Lindqvist, P., Johansson, 24 27 4,523-538. (4), 2,185-189. (2), nentoa eiwo Psychiatry of Review International , , 51 , 380 67 , Pain 5,413-418. (5), 73 2,271-279. (2), 1,107-113. (1), β- 4,285-292. (4), , nopi eesi h eta emna rai h aertbrain. rat male the in area tegmental ventral the in levels endorphin 114 , 59 12,62-70. (1-2), 63-69. , urn pno nEdcieadMtblcResearch Metabolic and Endocrine in Opinion Current h mrcnjunlo addictions on journal American The , 9 , 7,56-62. (7), 77 3,517-523. (3), Addiction ri Research Brain , , 30 13 hsooy&Behavior & Physiology 5,107-116. (5), 3,231-251. (3), 20 , 104 , 1) 1966-1978. (12), 251 , 2,380-383. (2), 28 4,246-261. (4), , 54 rgadachldependence alcohol and Drug hraooyBohmsr and Biochemistry Pharmacology 1,45-53. (1), , 6 54-59. , ersineresearch Neuroscience norn reviews Endocrine h ora of Journal The ri research Brain Neuroen- Phar- Brain , The 109 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. un .(05.Eegneo e ieecsi h eeomn fsbtneueadaueduring abuse and use substance of development the in differences mor- sex in of differences Emergence sex manipula- of hormone adolescence. Reversal (2015). neonatal by C. (2002). rats J. Kuhn, in R. gray Bodnar, periaqueductal & ventrolateral W., the tions. from D. peri- elicited Pfaff, ventrolateral analgesia S., rats. the phine Ogawa, in from differences K., elicited gonadectomy E. antinociception protein and Krzanowska, Morphine receptor sex opioid to (1999). on sensitive J. hormones is R. gray gonadal Bodnar, Reward aqueductal of & role Mesolimbic K., The in E. (2008). Estradiol Krzanowska, P. of S. Welch, Role rats. & arthritic the L., in and V. density Haller, Differences C., Sex Addiction. M. Opiate Kren, (2020). and I. Cocaine area to preoptic L. Vulnerability intra-medial Perrotti, and of & Circuits properties S., Affective S. (1999). Kokane, adolescent M. rats. G. with male are Alexander, in associated analgesia & Factors testosterone G., of morphine (1999). M. injections Packard, supraspinal F. E., in Nyberg, B. & differences King, steroids. L., anabolic-androgenic Sex Berglund, agents: G., doping (1999). D. of use Isacson, S. chronic M., and J. A. acute Mogil, Kindlundh, after & thermoregulation G., in genotype. S. on differences dependent Wilson, Sex gonadec- B., gender, (2000). Kest, of E. Roles Hopkins, mice. (1989). & in rats. administration J. M., in morphine R. Adler, morphine Bodnar, intracerebroventricular B., & of Kest, L., effects M. analgesic Cooper, the Behavior M., in and J. Biochemistry phase Kiefel, estrous B., and insular Kest, tomy of L., responses K. opiate-induced Kepler, in differences day-night triangularis. and maniculatus Sex and Peromyscus 477-482. sex (1987). mice, of G. Effects deer IV. D. wild Innes, morphine: of & hypothe- actions M., addiction the Kavaliers, steroid on influences anabolic Endocrine An (1984). R. hormones?: strain. George, on & Hooked G., B. Kasson, (1989). adult D. in H. conditioning Kleber, place & sis. induced B., sex-dependently K. Morphine Kashkin, Sex (2008). R. (2015). N. M. cohort rats. D. Zarrindast, Wistar population-based Juurlink, & a & M., therapy: M., opioid Karami, M. chronic during Mamdani, death A., overdose I. use and study. Dhalla, steroid escalation X., dose anabolic-androgenic Camacho, in of differences T., Associations Gomes, E., (2018). Kaplovitch, graphs. I. acyclic J. directed using Hudson, analysis & an 2601-2608. G., disorders: behavioral H. other steroid Jr, with anabolic-androgenic Pope with G., nonusers. men AAS Kanayama, of with Features and (2009b). users G. AAS H. nondependent dence Jr, with use Pope comparison & a steroid I., anabolic-androgenic dependence: J. Past Hudson, (2003). G., problem?. Kanayama, G. underrecognized H. an Pope, treatment: & abuse D., substance psychiatry for R. Weiss, admitted men H., among G. Cohane, G., Kanayama, Jama ri research Brain , lSone PloS iesciences Life 102 , , 262 13,130-137. (1-3), 64 uoenjunlo pharmacology of journal European hraooy&therapeutics & Pharmacology 2,156-160. (2), 2) 3166-3170. (22), , 10 , 8,e0134550. (8), , 34 929 ora fPamclg n xeietlTherapeutics Experimental and Pharmacology of Journal uoenjunlo pharmacology of journal European 1) 1627-1634. (17), 1,1-9. (1), , 34 1,119-127. (1), ersineletters Neuroscience ersineletters Neuroscience , 153 , 55-78. , 582 Addiction 21 13,78-87. (1-3), , hraooyBohmsr n Behavior and Biochemistry Pharmacology 291 , , 2,126-128. (2), 269 578 rnir nBhvoa Neuroscience Behavioral in Frontiers , 94 3,149-152. (3), 23,177-184. (2-3), 4,543-553. (4), ri research Brain scooia Medicine Psychological , rgadachldepen- alcohol and Drug 289 h ora fclinical of Journal The , 3,1370-1375. (3), 821 1,224-230. (1), Pharmacology , , 48 , 27 14 (15), (3), . Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. hla,E,Mree .G,Tci . oe . civn,S,&Taae .(05.Eet fanabolic- of Effects (2015). L. Trabace, & S., Schiavone, M., function. Bove, reward P., Tucci, brain modify on G., steroids androgens Androgenic-anabolic M. (1995). Morgese, E. E., of R. Mhillaj, Harlan, safety & and P., G. Efficacy Dohanich, (2010). J., T. G. β- Hebert, Guyatt, S., & C. Menard, N., H. in Catalano, review. differences systematic M., gender a Manzotti, 1150. and alopecia: C., androgenetic Sex for M. (2018). therapy F. Perret, finasteride S. M., Greenfield, J. & Mella, E., D. Sugarman, disorders. R., use V. substance Votaw, dependence hydrochloride K., R. of to McHugh, cases Three response use. (1996). steroid in (2016). T. anabolic A. hippocampus Petersen, T. with & rat Milner, associated K., & the Williamson, . in J., . A. . receptors A., McBride, opioid T. Kempen, delta Van stress. of D., chronic A. distribution and Dyer, K., acute subcellular Stafford, in C., S. differences Odell, in Sex S., self-administration B. Hall, (2017). S., E. Mazid, Chartoff, & rats. in D., female A Potter, and dynorphin S., male decanoate. of Page, conversion nandrolone M., Pravetoni, Enzymatic of M., administration (2007). Mavrikaki, by F. affected Nyberg, is & brain J., rat Bergquist, the M., determined brain Hallberg, rat K., the Magnusson, in receptors peptide opioid kappa autoradiography. of by density the affects dose-dependently nistration Bergstr C., hypotha- Birgner, of K., steroids. characteristics Magnusson, gonadal binding the by of rats Modulation in (1991). biology receptors F. self-administered Piva, opioid intravenously & of mu D., acquisition lamic Dondi, the P., Limonta, in R., differences Maggi, Sex rats. (1999). in E. heroin M. and Carroll, in cocaine & differences analgesia. J., morphine Sex W. in Lynch, differences (2008). sex Z. eliciting for A. essential Murphy, are neuroscience & gray periaqueductal analgesia. X., midbrain opioid Wang, rat in R., dimorphism sexual D. Opioid of Loyd, of neuroanatomy Models The Animal (2014). in Z. neurology A. Differences Murphy, mental Sex & receptor R., (2020). Androgen D. G. Loyd, P. (2016). Mermelstein, hypotha- Y. & of ontogenesis M., J. postnatal Reward. Esguerra, Ro, and M., Testosterone & N. (1991). K., Lopresti, F. M. Piva, & Chung, R., S., Maggi, lamic Q. D., Dondi, Auh, P., J., Limonta, Asgar, opioid Y., among interactions Zhang, regulates addiction: transcriptionally S., and analgesia K. of opioid effects in Lee, differences The Sex receptors. (2016). (2013). estrogen K. A. and Ho, M. receptors & S., rats. Smith, W. in & C. self-administration M., Lee, heroin A. and Robinson, cocaine on A., contact M. social , Feinstein, and C., cycle, estrous J. sex, Strickland, T., R. Lacy, 233 nopi muoeciiyi h a brain. rat the in immunoreactivity endorphin 1) 3201-3210. (17), μ , [H iyrmrhn)oii eetr nterat. the in receptors opioid ) ([3H] urn eulHat Reports Health Sexual Current 40 13,113-121. (1-3), , 28 , 5) 14007-14017. (52), 259 Neuropeptides Psychopharmacology 57-63. , lnclpyhlg review psychology Clinical erbooyo Stress of Neurobiology μ- Psychopharmacology podrcpo xrsini a rgmnlganglia. trigeminal rat in expression receptor opioid oeua pain Molecular , m . yeg . aleg .(09.Nnrln eaot admi- decanoate Nandrolone (2009). M. Hallberg, & F., Nyberg, L., ¨ om, 43 rnir nneuroscience in Frontiers rts ora fsot medicine sports of journal British 1-9. , 2,105-111. (2), , 234 ri research Brain , , , 6,977-987. (6), 5 144 9 , 37-53. , 22 66 1,45. (1), 1,77-82. (1), h ora fseodbohmsr n molecular and biochemistry steroid of Journal The 12-23. , eeomna ri research brain Developmental , , 669 9 rhvso dermatology of Archives 295. , Peptides 2,255-262. (2), μ- , 30 podrcpo xrsini the in expression receptor opioid 1,69-70. (1), , 28 Neuroscience 4,851-858. (4), Psychopharmacology , , 62 146 , 1,131-136. (1), 331 1) 1141- (10), ora of Journal 52-61. , Experi- Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. aqailo . iTr,R,Nbr,F,&Sapnt,S 20) onrglto fdlaoii receptor cells. opioid hybrid delta of neuronal Down-regulation in (2000). steroid S. anabolic Spampinato, an & F., by somatic Nyberg, mRNA spontaneous R., Toro, of Di expression A., dose-linked Pasquariello, morphine Gender-and mice. in (2006). rats. withdrawal A. in opiate threshold Contarino, pain & on F., testosterone Papaleo, of Role pharmacology (1995). intra-nucleus and K. of physiology V. properties of Mulgaonker, affective & Rewarding R., J. (1997). diagnosis, Padnekar, M. (OPIAD): G. testosterone. Alexander, deficiency of & androgen H., injections A. accumbens Opioid-induced Cornell, G., (2016). M. Packard, S. M. review. Wosnitzer, literature and & implica- management, steroids: K., differ- androgenic T. sex anabolic behavior. Do and O’Rourke, addictive opioids of between (2010). development Interactions clinical E. the (2012). and for Sarton, M. experimental tions & Hallberg, human & of L., F., meta-analysis Aarts, Nyberg, and T., review Stijnen, systematic A J., from for Zacny, analgesia? evidence studies. opioid B., of in Retrieved Lack Kest, exist (2001). A., ences D. Dahan, B. adminis- M., Fischer, steroid & Niesters, anabolic–androgenic D., high-dose Use. J. following Jr, monkeys Wines rhesus Substance G., in tration. Kanayama, dependence G., in or H. tolerance antinociception Jr, opioid opioid Pope on Differences treatments S., hormone S. steroid Negus, Gender gonadal of Effects monkeys. and (2002). rhesus K. ovariectomized N. in Mello, Sex & S., S. Negus, 24 in September syndrome 2020, 28. withdrawal on morphine in-substance-use May on testosterone https://www.drugabuse.gov/publications/research-reports/substance-use-in-women/sex-gender-differences- of 2020, Effect (2008). NIDA. H. Rezazadeh, & Modulatory M., rats. antinociception: R. mediated A. opioid and Nayebi, pain in differences dysreg- hormones. Sex in gonadal (2019). dynorphin of A. and E. role Afify, CREB & accumbens A., nucleus S. of Nasser, study. Roles microdialysis (2013). A. a motivation. W. of Carlezon, rats: ulation & tolerant W., morphine hormones J. gonadal Muschamp, in of role accumbens and nucleus differences pharmacology Sex the of (2007). in journal M. level Jorjani, & glutamate F., on Kobarfard, B., Shafaghi, has Z., Mousavi, position Intrauterine (2003). G. Laviola, 386-400. patients. (3), & opioid-dependent Vigan`o, brain D., in on D., influence Parolaro, Preston, craving long-term & P., and Palanza, stress . S., . . life Morley-Fletcher, abuse M., daily alcohol Mezghanni, L., in and J. drug differences Lin, of Sex M., Vahabzadeh, journal anterior A., (2018). pituitary K. rat Phillips, L. of J., K. Response W. litera- Kowalczyk, environment. (1986). M., biased M. steroid L. a B. gonadal Moran, Cox, in of & changes G., evidence to J. products emerging Rosenberger, prodynorphin H., lobe processing: A. Hassen, pain J., in C. Molineaux, differences sex Qualitative (2020). ture. S. J. Mogil, sa ora fandrology of journal Asian aueRvesNeuroscience Reviews Nature Psychoneuroendocrinology Pain , 151 1,61-68. (1), odSrn abrprpcie nmedicine in perspectives Harbor Spring Cold , 554 iesciences Life eaiua ri research brain Behavioural μ- podrcpo est n eaiu nmice. in behaviour and density receptor opioid 23,145-149. (2-3), , , 39 10 , Psychopharmacology , 44 1-13. , urn rlg reports urology Current 26 423-424. , 5,765-769. (5), eairlneuroscience Behavioral 5,512-523. (5), 8,789-796. (8), , 237 116926. , n e Neurobiol Rev Int 23 , Neuroreport 170 , 159 , 1,110-118. (1), 17 3,275-283. (3), , 111 1) 76. (10), , , , 1,219. (1), 102 3 11 2,a012005. (2), Endocrinology 4,863-867. (4), 189-206. , Psychoneuroendocrinology , 119 5,2297-2305. (5), h American The ninjournal Indian European , 28 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. n yAeadSx—Uie tts 0821.MW obMra kyRp2020;69:298–302. Rep Wkly Mortal Morb MMWR Dispens- Prescription 2008–2018. Opioid A. Outpatient States, Dahan, Adult United & in — Variation JL. Sex . Losby . and . P, DOI: Age Seth D., by Jr, Nieuwenhuijs, GP ing B., volunteers. Guy healthy Anesthesiologists Kest, LZ, in of J., Schieber Society study R. American experimental Hartigh, A. the analgesiaan den Dehpour, of morphine Journal & R., in The differences Romberg, . Sex . mice. E., . female (2000). B., Olofsen, and Sharif, E., male J., Sarton, gonadectomized M. Paydar, in M., dependence Behavior Ghasemi, morphine-induced and K., Biochemistry of to D. response Study Anaraki, the S., modify (2009). androgen Sianati, and M., estrogen Sadeghi, Both (2016). M. S. Moenter, , & Report and L., neurokinin-3 matter? L. of activation it Burger, Does A., K. care. substitution. Ruka, palliative hormone in with hypogonadism treated Opioid-induced patients (2009). four Z. on Zylicz, & E., Roantree, brain alters morphine to rats. exposure in Prenatal istics (1995). I. Vathy, & Riman´oczy, A., receptors. pharmacology.2 Opiate opioid (1995). kappa T. in Reisine, anabolic-androgenic differences Sex on Dependence (2011). Y. testosterone 2-16. L. (2014). (1-2), of Liu-Chen, A. role & Dhawan, K., review. The & brief Rasakham, and D., (2017). report R. C. case Pattanayak, T. a A., steroids: Hsieh, Parmar, & R., J., analysis. Ranjan, T. pilot Furnish, retrospective D., a in Sisul, health hypogonadism: differences men’s H., opioid-induced strain in and S. therapy Sex Patel, supplemental R. (1985). A., A. A. O. J. Genazzani, SHRs. Wojcechowskyj, Raheem, and & & WKYs J., in Barlament, effects . . K., temperature . L. morphine-induced Vaughn, A., M., Cubeddu, R. S., Quock, rats. Merlini, gonadectomized the to A., plasma in administration and receptors Giannini, estradiol brain opioid and E., in mu differences Casarosa, of Sex Decrease F., (2009). rat. Ninni, (1987). male L. aged N., Martini, the Pluchino, of & hypothalamus D., the Dondi, in P., and Limonta, brain R., the Maggi, modulates F., Piva, Nandrolone (2003). dimorphism. sexual M. of Vlaskovska, role Biochemistry & tolerance/dependence: and L., and Kasakov, analgesia M opioid E., and & Pavlova, non-opioid R., T., plasma Ivanova, A. and T., Genazzani, pituitary Philipova, E., on A. replacement Panerai, steroid rat. D., gonadal the Cocchi, and in gonadectomy A., potential of and V. Effect tolerance, Locatelli, overdose, (1982). A., hamsters: Penalva, in gonadal F., dependence Petraglia, of Androgen Role (2004). I. (2011). R. mechanisms. R. Wood, J. & Traynor, D., & K. Peters, B., J. rats. Becker, Sprague–Dawley GTP E., female S] Jutkiewicz, and [35 M., mu-opioid-stimulated S. on Graves, hormones M., E. Peckham, 157 http://dx.doi.org/10.15585/mmwr.mm6911a5 2,752-763. (2), Endocrinology , ri research Brain 11 4,1208-1213. (4), , 111 5,429-436. (5), Neuroscience , 111 , , 91 κ- 690 4,1224-1229. (4), podrcposi rut isetnnuosfo aemice. male from neurons arcuate in receptors opioid 4,604-609. (4), Psychopharmacology 2,245-248. (2), Neuropharmacology,34 , 130 eh scityJ Psychiatry Delhi β- 4,971-981. (4), nopi otn olwn etseoe dihydrotestosterone testosterone, following content endorphin γ idn n opiemdae nioieto nmale in antinociception morphine-mediated and binding S dacsi alaieMedicine Palliative in Advances 24 iesciences Life Neuroendocrinology , 218 , 93 , ri eerhbulletin research Brain 5,4342 doi:10.1016/0028-3908(95)00025- 463-472. (5), 3,483-492. (3), 17 5,1245-1254. (5), 2,481-4. (2), , 40 4,391-398. (4), , 89 4,411-423. (4), μ podrcpo character- receptor opioid , 14 rhvso Physiology of Archives , 8 mrcnjunlof journal American 4,323-326. (4), 2,69-74. (2), β- iesciences Life nopi levels endorphin Anesthesiology: Endocrinology Pharmacology le,E E. E. uller, ¨ , 88 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. enr .M,Bret .C,Co,C . ikr .J 20) e ieecsi (-)-pentazocine in differences nociceptive Sex thermal a (2003). using J. strains M. rat Picker, four & in D., C. and Cook, morphine C., to in A. comparison variation Barrett, Menstrual antinociception: leu- M., (2010). hormone, S. pitu- J. Lautenbacher, releasing Terner, hormones. and & gonadal thyrotropin K., brain with of Schepelmann, correlation rat H., distribution pain: Vedder, the experimental regional M., in Peters, The M., Teepker, and (1991). re- Y. opioid P, S. brain itary. substance W. rat met-enkephalin, of Man, enkephalin, responses & Regional F., Tang, (1990). vs. Z. indices Ota, replacement. reproductive & testosterone of pharmacology and M., modulation castration Asanuma, Testosterone upon N., ceptors (2006). Ogawa, M. H., R. Takayama, modulation Craft, rats. hormone & male Gonadal in C., (2005). antinociception M. Ulibarri, morphine rats. R. E., female Craft, and & J. male C., in Sumner, K. antinociception Rice, nociception, opioid E., delta of J. and modulation Folk, kappa, M., hormone mu, of rats. C. steroid Ulibarri, female Gonadal C., and (2003). E. male M. Stoffel, in of R. indices expression Craft, reproductive and & and development M., antinociception the C. morphine rats. on Ulibarri, female hormones C., and gonadal E. male differences of Stoffel, in effects Sex-related morphine The of effects (1999). (2001). stimulant D. the T. the Rodaros, in M. & morphine J., Smith, clearance. Stewart, of morphine & infusion via E., intravenous testosterone L. chronic of Mather, Therapeutics following role M., development modulatory exercise Lau, tolerance to rat: access W., and free A. with antinociception Wright, rats in in M., opioids S. of effects South, the to female Sensitivity and (2003). L. male D. by Yancey, wheels: & and A., exposure M. morphine Smith, prenatal rats. by gonadal adult altered and in are steroids exposure gonadal structures morphine brain prenatal seizure-controlling by in altered is Riman´oczy, rats Slamberov´a, R., female and of male Density adult (2003). treatment. anabolic- I. of hormone Vathy, to hippocampus & addiction J., of the C. feature in Schindler, a N., as Bar, use Riman´oczy,Slamberov´a, A., Multisubstance R., (2009). I. Engstrom, steroids. & androgenic F., Nyberg, K., Skarberg, regulation. hormonal periventricular anteroventral and the differentiation in 473-484. sexual expression (6), gene rat: proenkephalin the heptadecapep- and of opioid Prodynorphin nucleus the (1991). for B. in pathway R. conversion enzyme Simerly, in A-converting distinct Testosterone of of to Characterization related Role (1992). endoprotease tide?. F. An (2019). Nyberg, cord. N. & spinal H. E., human Yadav, M. Castello, & J., K., Silberring, J. Gupta, A., 675-681. Goyal, (4), Rats. K., in Analgesia N. Exercise-induced Singh, Swimming use. K., opioid prescription D. in differences Sharma, Gender (2017). B. psychiatry L. Cottler, in & opinion W., C. Striley, M., Serdarevic, ˇ ˇ Neuropeptides ora fBooia Chemistry Biological of Journal μ- podtlrneadpyia dependence. physical and tolerance opioid , , 297 70 3,355-358. (3), 1,446-457. (1), uoenAdcinResearch Addiction European Hippocampus , , 19 30 4,287-292. (4), 4,238. (4), . ie,M . cide,C . ah,I 20) uoii receptors Mu-opioid (2002). I. Vathy, & J., C. Schindler, A., M. Riley, A., ´ ri eerhbulletin research Brain , 13 , 4,461-471. (4), iesciences Life 267 3) 21324-21328. (30), ninjunlo hraetcleuainadresearch and education pharmaceutical of journal Indian , 15 25 Psychopharmacology , , 79 eerhcmuiain nceia ahlg and pathology chemical in communications Research 2,99-106. (2), Neuropsychobiology 58 2) 2119-2127. (22), eaiua ri research brain Behavioural 4,391-400. (4), ora fPamclg n Experimental and Pharmacology of Journal oeua n ellrNeuroscience Cellular and Molecular h ora fPain of Journal The , 168 , Pain 61 4,426-434. (4), 3,131-140. (3), , 103 , 102 3,285-302. (3), μ oii receptors -opioid , 12,89-98. (1-2), 6 4,261-274. (4), Current , , 53 2 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. isnN ais ,St ,SihHI,DvsN.Du n podIvle Over- Opioid-Involved and 2020;69:290–297. Drug Rep NL. Wkly immunore- receptor Mortal Davis opioid delta in Morb differences IV, Sex MMWR (2002). H J. amygdala. A. medial McDonald, rat Smith & in F., activity 2017–2018. Mascagni, P, A., M. States, Wilson, Seth United M, — DOI: Deaths Kariisa dose delta N, of hippocampus. regulation rat Hormonal Wilson the (2011). in A. memory cells T. have and pyramidal Milner, learning and rats & interneurons of female D., in J. not immunoreactivity Chapleau, receptor but opioid A., male Torres-Reveron, Periadolescent J., T. rats. Williams, (2008). adult G. do S. than morphine Holtzman, to the & , response in C., in distribution activity C. opioid motor dimorphic Michaels, higher Sexually A., (1986). D. J. White, steroids. S. gonadal Wiegand, by & manipulation E., G. area: in Hoffmann, preoptic E., steroids R. androgenic Jr, Watson of anabolic Regulation with doping (1986). α- of L. effects S. Wardlaw, Adverse bodybuilding. and (2018). sports the https://doi.org/10.23736/S0391-1977.18.02810-9 E. recreational in Nieschlag, behavior athletics, abstinence competitive & attenuates E., and Vorona, tolerance morphine of development and 5 the & rat. a male prevents Finasteride, in . morphine, (2007). . . behavior A. of Ahmadiani, E., locomotor & G. and hormones: J., Verdi, sex reward, Torres, to P., release, exposure dopamine Jara, & early rats. mouse accumbens C., female by the morphine-induced . Bonansco, neurons . in . on dopaminergic J., Mart´ınez-Pinto, levels S., effects of peptide Ogawa, A., Programming opioid C., G. (2019). M. influence Sotomayor-Z´arate, R. Zamorano, Tsuda, expression B., L., receptor J. Vel´asquez, estrogen V. Spencer-Segal, and pathway. D., Sex fiber A. mossy (2013). Gonzalez, hippocampal and A. S., nociception T. on Kahlid, effects A., Milner, steroid T. Anabolic–androgenic Kempen, (2011). Van the M. in R. rats. Craft, dopamine male & accumbens in I., nucleus antinociception R. morphine and Wood, Testosterone T., (2008). K. I. Tsutsui, R. Wood, & hamster. M., Syrian S. male Sato, use. sexually L., steroid is J. anabolic Triemstra, of circuitry manifestations modulating Behavioural pain (2005). W. Brainstem 571-595. G. (7), function. Currier, (2000). receptor & L. J., opioid kappa H. A. Trenton, of and Fields, doses mu & low to M., of respect the Influence J. with on dimorphic Mitchell, (2006). strains. J. gonadectomy A., four M. S. of of Picker, rats Tershner, & Influence female S., and male S. in (2002). Negus, tolerance M., morphine J. , L. and M. antinociception Lomas, morphine C., Picker, on A. naltrexone & Barrett, rats. M., E., female J. and Grossell, Terner, male in C., opioids A. of effects Barrett, antinociceptive M., J. Terner, assay. 89 122 eaorpnSiuaigHroei h yohlmsb Testosterone. by Hypothalamus the in Hormone Stimulating Melanotropin omnsadBehavior and Hormones 2,188-199. (2), http://dx.doi.org/10.15585/mmwr.mm6911a4 12,90-101. (1-2), eaiua pharmacology Behavioural rnir nPharmacology in Frontiers Psychoneuroendocrinology , 95 , 2,206-220. (2), 51 ersineletters Neuroscience , 5,605-610. (5), 14 ersineletters Neuroscience 1,77-85. (1), hraooyBohmsr n Behavior and Biochemistry Pharmacology , β- 10 nopi,CriorpnLk nemdaeLb etd,and Peptide, Lobe Intermediate Corticotropin-Like Endorphin, 295. , , ri research Brain 33 α- 26 euts niio,ptnitsatnccpieeffects antinociceptive potentiates inhibitor, reductase 3,386-394. (3), , 328 , 552 Psychopharmacology 2,160-164. (2), 66-70. , iev endocrinologica Minerva , hraooyBohmsr n Behavior and Biochemistry Pharmacology 398 Pain 1,157-163. (1), , 85 Endocrinology 12,153-159. (1-2), , 163 , 99 2,183-193. (2), 3,500-508. (3), , 43 , N drugs CNS 119 4,476-488. (4), Neurobiology 1,19-24. (1), Pain , 19 Posted on Authorea 2 Oct 2020 — The copyright holder is the author/funder. All rights reserved. No reuse without permission. — https://doi.org/10.22541/au.160167496.62768848 — This a preprint and has not been peer reviewed. Data may be preliminary. hn,X,Zag . sa,J,Nu .Y,Le . e,K . o .Y 21) e differences Sex (2014). Y. J. Ro, & . . . S., K. Lee, J., Lee, Y., K. males Niu, in pain effects J., stimulus Asgar, and on Y., in stimulus Gender cold-water Zhang, a (2007). X., of L. Zhang, effects Lu, The & (2004). J. females. N. . . and Beckman, . & H., P., Qin, J. Zacny, J., by Shi, users. regulation Y., heroin Fang, abstinent 485-492. pituitary: in H., (3), anterior responses D. rat cue-induced Epstein, in the difference S., in Zhang, system J., enkephalin Yu, The drugs. psychotropic (1983). and hormones S. the steroid J. in gonadal weights Hong, Influence testicular & and (1999). K., body H. Yoshikawa, and Kelestimur, levels, & testosterone R., FSH, M. rat. LH, Gezen, male serum S., developing on Canpolat, exposure S., male morphine Sandal, in chronic S., stimulus of Kutlu, discriminative V., a Konar, as B., Testosterone Yilmaz, (2011). E. intra- Antzoulatos, & reinforcement: V., Testosterone rats. N. Vertelkina, (2004). I., W. R. D. Wood, Self, & hamsters. C., and Schad, rats 298-305. L., male (3), in Chu, self-administration R., intracerebroventricular and L. venous Johnson, I., humans. R. and Wood, animals from Insights dependence? steroid anabolic neuroendocrinology Anabolic–androgenic in in attraction?. (2008). dependence fatal I. a hydrochloride R. Wood, steroids: Nalbuphine Anabolic (1999). (2006). I. S. R. Lukas, Wood, & H., Pope, users. A., steroid Gruber, J., Wines, μ oii eetrepeso ntieia agi ne ystscniini rats. in condition myositis a under ganglia trigeminal in expression receptor -opioid , hraooyBohmsr n Behavior and Biochemistry Pharmacology 18 2,151-161. (2), hraooyBohmsr n Behavior and Biochemistry Pharmacology mrcnJunlo Addictions on Journal American rhvso andrology of Archives , 29 4,490-506. (4), , 43 , , 3,189-196. (3), 8 100 Endocrinology 2,161-164. (2), 27 1,185-190. (1), , 78 ora fneuroendocrinology of Journal 4,653-659. (4), hraooyBohmsr n Behavior and Biochemistry Pharmacology , 113 4,1218-1227. (4), Psychopharmacology uoenjunlof journal European , 18 3,227-228. (3), Frontiers , , 171 86