<<

58 2

m c evans and g m anderson Nutritional regulation 58: 2 R107–R128 Review of

Neuroendocrine integration of nutritional signals on reproduction

Correspondence Maggie C Evans and Greg M Anderson should be addressed to M C Evans Centre for Neuroendocrinology and Department of Anatomy, University of Otago School of Email Medical Sciences, Dunedin, New Zealand maggie.evans@anatomy. otago.ac.nz

Abstract

Reproductive function in is energetically costly and therefore tightly regulated Key Words by nutritional status. To enable this integration of metabolic and reproductive function, ff information regarding peripheral nutritional status must be relayed centrally to the ff -releasing hormone (GNRH) that drive reproductive function. ff The metabolically relevant hormones leptin, insulin and ghrelin have been identified ff GNRH as key mediators of this ‘metabolic control of fertility’. However, the neural circuitry ff fertility through which they act to exert their control over GNRH drive remains incompletely understood. With the advent of Cre-LoxP technology, it has become possible to perform targeted -deletion and gene-rescue experiments and thus test the functional requirement and sufficiency, respectively, of discrete hormone– signaling pathways in the metabolic control of reproductive function. This review discusses the findings from these investigations, and attempts to put them in context with what is known from clinical situations and wild-type animal models. What emerges from this discussion is clear evidence that the integration of nutritional signals on reproduction

Journal of Molecular Endocrinology is complex and highly redundant, and therefore, surprisingly difficult to perturb. Consequently, the deletion of individual hormone–neuron signaling pathways often fails to cause reproductive , despite strong evidence that the targeted pathway plays a role under normal physiological conditions. Although transgenic studies rarely reveal a critical role for discrete signaling pathways, they nevertheless prove to be a good

strategy for identifying whether a targeted pathway is absolutely required, critically Journal of Molecular involved, sufficient or dispensable in the metabolic control of fertility. Endocrinology (2017) 58, R107–R128

Introduction

Mammals, particularly females, must invest an enormous receive and integrate information about peripheral amount of energy into conceiving, carrying and rearing metabolic status. Indeed, each level of the HPG axis has their progeny to preserve their genetic lineage. Therefore, the capacity to respond to changing levels of nutritional to ensure adequate energy is available to support the cues (Comninos et al. 2014). However, the gonadotropin- high demands of pregnancy and successful weaning releasing hormone (GNRH) neurons residing in the of offspring, reproductive function tends to become are the primary drivers of reproductive suppressed during times of low-energy availability. For function, and impaired GNRH signaling is considered this coordinated regulation of energy homeostasis and the underlying cause of reproductive suppression due to fertility to occur, the hypothalamic–pituitary–gonadal nutritional stress. (HPG) axis governing reproductive function must

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access

10.1530/JME-16-0212 Journal of Molecular Endocrinology safeguard reproductive function and thus ’ survival. safeguard reproductive function and thus species’ survival. the complexityandinherent redundanciesbuiltinto relayed centrallytotheGNRH neuronsandalsohighlights important insightintohow peripheral metabolicstatusis mechanisms underlying nutritional infertility provides in isolation genetic deficiencies,asnosinglepathwayiseveroperating meaningful approach,exceptperhapsincasesofspecific and is arguably not anexperimentallyandphysiologically exerts itsimpactonGNRHreleaseprovesextremelydifficult the specific pathways bywhicheachfactorindependently due totheneteffectofallthese.Trying todisentangle suppression of the reproductive neuroendocrine axis is response) isinitiated,anditseemslikelytheconsequent (as wellasactivationofthestressaxesandcounter-regulatory multitude ofgut-andadipose-derivedhormoneresponses or chronicunder-nutrition,aseriesofeventsinvolving function isextensive. from thesesignalsandthengoontomodulateHPGaxis extra-hypothalamic) circuitry abletoreceiveinformation energy statuscentrallybutalsothehypothalamic(and nutritional signalsrelayinginformationaboutperipheral challenging areaofresearch. Notonlyaretherenumerous and nutritionalsignalsinvolvedremainsanactive hypothalamus, unraveling the specific neuralsubstrates reproductive functionisknowntooccurprimarilyinthe 2003 availability appearstobenonlinear( 2007 related totheseverityofenergeticstress( Furthermore, thesuppressionofGNRH/LHpulsatilityis Nosbisch 1991 to immediatechangesinenergyavailability( from long-term changes in adiposity and is largely due function duetonutritionalstresscanoccurindependently 2011 unquestionably associatedwithinfertility( 2005 hypogonadism ( the ,ultimatelyresultinginhypothalamic secretionof suppressed, whichinturnreducespituitary periods ofenergeticstress,GNRHpulsesbecome & Moenter2010 which thengoontodrivegonadalfunction( hormone (LH) and follicle-stimulating hormone (FSH)) stimulates thereleaseofgonadotropins(luteinizing glandwhereit and travelstotheanteriorpituitary pulsatile mannerintothehypophysealportalcirculation DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review In response to an energetic stress, such as an acute fast In responsetoanenergeticstress,suchasacutefast Under normalconditions,GNRHissecretedina ). Althoughthisdelicatecontrolofmetabolicand ); yet,thedependenceofLHpulsatilityonenergy ), suppressionofneuroendocrinereproductive , Huang in vivo t al et , Compagnucci Loucks , . Nevertheless, identifying the discrete . Nevertheless,identifyingthediscrete . 2008 Herbison 2016 et al ). Althoughhuman obesity is . 1998 et al m

, . 2002 c © 2017SocietyforEndocrinology

Wade &Jones2004 evans ). However, during Loucks &Thuma and , Castellano Printed inGreatBritain De Souza Souter g Cameron &

m

Christian anderson t al et et al et al ). . . . reduced levelsofplasmaglucose,suppressedthyroid limited to)alteredlevelsofgutandadiposehormones, endocrine-metabolic aberrations, including (but not hypothalamic amenorrhea(FHA))exhibitarangeof presenting withnutritionalinfertility(i.e.functional of reproductive function is challenging. Clinically, women specific componentscriticallyinvolvedinthesuppression nutritional stressismultifaceted,soidentifyingthe As mentionedpreviously, thebody’s responseto Identifying thekey players normal metaboliccontroloffertility. for, criticallyinvolvedinand/orsufficienttopermitthe of whetheridentifiedpathwaysareabsolutelyrequired inherent redundanciesofthesystemthroughdiscussions modulate reproduction,whilealsoemphasizingthe neuroendocrine pathwayswherebynutritionalsignals This reviewwillattempttohighlightsomeofthekey to nutritionalstress.For example,preventingthe is thoughttomediatereproductive suppressiondue in response to complete fasting ( in responsetochanges fatmass,butfallrapidly leptin concentrationschangegraduallyovertime feeding andincreaseenergyexpenditure.Serum sufficiency andacts onthe hypothalamus tosuppress to bodyfatness,andsignalsenergy proportion in is secreted fromadipocytesintothebloodstream appears tobeaparticularlyimportantfactor. Leptin chronically elevatedcortisolconcentrations( function, alteredgrowthhormonesecretionpatternsand Chan Leptin, thehormoneproductofobesitygene( Leptin energy availability. axes activationinreproductivesuppressionduetolow function andwillalsobrieflytouchupontheroleofstress whereby these key nutritional factors influence HPG review will therefore focus on the discrete mechanisms that inatleastsomecasestheireffectsareadditive.This GNRH/LH pulsatility(andthusfertility)thanothersand insulin andghrelin,exertamorecriticalinfluenceover studies thatcertainnutritionalsignals,suchasleptin, reproductive axis,weknowfrombothanimalandclinical factors independentlysuppressestheneuroendocrine 2011 & Warren 2006 1989 of reproduction Nutritional regulation Published byBioscientifica Ltd. ). Although it remains a possibility that each of these , t al et Laughlin &Yen 1997 . 2003 , ), and this fall in leptin concentration Scheid &DeSouza2010

Downloaded fromBioscientifica.com at09/24/202102:55:05PM , Laughlin et al Boden 58 : . 1998 2 t al et , Berga Corr , Schneider . 1996 R108 t al et et al Ob via freeaccess ), , . .

Review m c evans and g m anderson Nutritional regulation 58:2 R109 of reproduction

starvation-induced fall in leptin with exogenous leptin was associated with elevated LH pulsatility and polycystic shown to prevent the starvation-induced delay in ovulation ovary syndrome (PCOS) (Laughlin et al. 1997), and insulin- in female mice (Ahima et al. 1996). Accordingly, women sensitizing drugs (e.g. metformin) have successfully with infertility due to a negative energy balance exhibit been used to induce ovulation and treat some of the reduced circulating leptin concentrations (Laughlin & Yen pathophysiological features associated with PCOS (Katsiki 1997, Corr et al. 2011), and administration of recombinant & Hatzitolios 2010). Encouragingly, animal studies human leptin to these women not only increased their LH support these clinical findings. For example, lean and pulsatility and restored ovulation but also improved their normoglycemic female mice exhibiting varying degrees thyroid and axes, independent from of insulin resistance and hyperinsulinemia were shown dietary changes or weight gain (Welt et al. 2004). These to have subtle alterations in their HPG axis and also had data suggest that an acute decrease in circulating leptin polyovular follicles (Nandi et al. 2010). Although insulin plays a key role in mediating the effects of low energy can act both peripherally and centrally to modulate HPG availability on reproductive function. axis function, we know that insulin’s central actions play In further support of leptin’s role in the metabolic a role in mediating many of its reproductive effects as regulation of fertility, humans and mice with a congenital mice exhibiting brain-specific deletion of insulin receptors leptin deficiency (ob/ob), as well as leptin (LepR)- (InsR) exhibit subfertility due to mild hypothalamic deficient db/db( ) mice, are infertile and obese despite being hypogonadism (Bruning et al. 2000). However, in contrast energy replete, and leptin treatment is sufficient to restore to these brain-specific InsR-knockout mice, our group body weight control and reproductive function in leptin- recently demonstrated that mice exhibiting forebrain deficient individuals (Ingalls et al. 1950, Hummel et al. neuron-specific deletion of InsR displayed normal HPG 1966, Barash et al. 1996, Chehab et al. 1997, Mounzih et al. axis function (Evans et al. 2015), which challenges the 1997, Farooqi et al. 1999, 2002). Like the global LepR- hypothesis that neuronal insulin signaling is required. deficient mice, mice exhibiting forebrain neuron-specific Nevertheless, many findings implicate abnormal insulin deletion of LepR are also infertile and obese, highlighting signaling in the pathogenesis of reproductive dysfunction. that leptin’s central actions, vs peripheral actions, are However, in contrast to leptin, central insulin signaling is critically involved in regulating body weight homeostasis not absolutely required for fertility. and fertility (Quennell et al. 2009). Lastly, central leptin immune neutralization using leptin antiserum caused Ghrelin a reduction in LH pulsatility in rats (Carro et al. 1997).

Journal of Molecular Endocrinology These findings, among others, convincingly demonstrate Unlike the anorexigenic insulin and leptin, that leptin plays an important physiological role in the which promote GNRH/LH pulsatility and reproductive regulation of metabolic and reproductive function. function, the orexigenic ghrelin has been shown to consistently suppress GNRH pulsatility and gonadotropin release (Furuta et al. 2001, Kluge et al. Insulin 2007, 2009, 2012, 2013, Kluge 2012). Although ghrelin The pancreatic insulin (best known for is predominantly produced in the stomach, ghrelin- regulating glucose homeostasis and body weight regulation expressing neurons have also been identified in the (Belgardt & Bruning 2010)) has also emerged as a key hypothalamus (Cowley et al. 2003), yet it remains unclear modulator of reproductive function. Clinically, diabetic whether the actions of brain-derived ghrelin are the same insulin insufficiency (i.e. type 1 diabetes) in humans is as those for circulating ghrelin. Under normal conditions, associated with delayed , reduced LH pulsatility and circulating ghrelin exhibits a circadian pattern whereby hypothalamic hypogonadism, despite sufficient energy its concentration becomes elevated in anticipation of availability; yet, improved insulin therapy has ameliorated meals. However, in women exhibiting chronic under- many of these issues (Codner et al. 2012). Additionally, nutrition, chronically elevated basal ghrelin levels are reduced insulin levels, presumably due to energy deficiency, observed and are correlated with suppressed reproductive are likewise associated with suppression of neuroendocrine function (Scheid & De Souza 2010). Ghrelin deficiency, reproductive function in women with FHA (Laughlin on the other hand, does not significantly affect fertility & Yen 1997), which further suggests insulin promotes or feeding behavior in mice (Sun et al. 2003). The latter GNRH/LH secretion. Conversely, hyperinsulinemia observation is likely due to the fact that the ghrelin and/or insulin resistance (i.e. type 2 diabetes), are often receptor (the growth hormone secretagogue receptor

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology However, therolesofthesemetabolic hormonesin effect ontheHPGaxis( inhibitory fertility andhasbeenshown to haveapredominantly adipocytes, is alsoimplicated inthemetabolic control of 2006 ( and appear to facilitate GNRH release and HPG function peptide YY3-36(PYY)arebothstimulatedbyfoodintake anorectic gut peptides -like peptide (GLP)-1 and and coworkers ( HPG axisfunction,asthoroughlyreviewedbyComninos are manyothernutritionalfactorsthatalsoinfluence relayed totheneuroendocrinereproductiveaxis,there major signalswherebyperipheral metabolic statusis Although leptin,insulinandghrelinappeartobethe Other nutritionalsignals suppresses theneuroendocrinereproductiveaxis. player inthemechanismwherebylowenergyavailability 2005 reduced leptin-inducedinhibitionofghrelin( Warren 2006 exhibit chronicallyelevatedghrelinlevels( and womenwithdisorderedeatingamenorrheaalso in theirnormallycyclingpeers( that areapproximately85%greaterthanthoseobserved women withamenorrheaexhibitplasmaghrelinlevels deficient mice( signaling improvesthereproductivephenotypeofleptin- on fertility. Insupportofthis,inhibitionghrelin leptin’s andinsulin’s effects, thereby acting as a‘brake’ accompanies nutritionalstressisthoughttoantagonize the increaseincirculating ghrelinconcentrationsthat Nevertheless, undernormalphysiologicalconditions, reproductive parameterswerenotassessedinthesemice. likely duetocompensationbyotherfactors.Unfortunately, mice, butfailedtocauselong-termimpacts( caused anacuteincreaseinfoodintakeandbodyweight induced reductioninLHsecretion. ghrelin-knockout mice were protected against a fasting- have beenmoreinformativetodeterminewhetherthe knockout miceisperhapsnotsurprising.Itwouldarguably intheghrelin- the lackoffertilityphenotypeobserved Considering ghrelin’s actionsonGNRHrelease, inhibitory of ghrelinpeptide, ghrelin signaling is not abolished. 2003 type 1a(GHSR1a))has constitutive activity( Beak DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review Chronic overexpression of ghrelin, on the other hand, ). Thesestudiesstronglyimplicateghrelinasakey ). , a hormone secreted by white , t al et Holst &Schwartz2004 . 1998 , Schneider Zhu Comninos , et al MacLusky et al . 2013 et al . 2008 t al et ), soevenintheabsence ). Furthermore,exercising . 2014 m

De Souza ), whichmaybedueto c © 2017SocietyforEndocrinology

. 2000 evans Klenke ). For example, the and Printed inGreatBritain , Qi t al et t al et Pinilla Schneider & g et al

Holst m Kalra

anderson . 2004 . 2014 . 2015 t al et t al et et al ), ). ), . . . peripheral signalsofnutritionalstatus( influence ofleptin,insulinandghrelin,aswellother is primarilythroughthisspecializednetworkthatthe neurons (termedtheGNRHneuronalnetwork)andit regulated by a network of hormone-sensitive afferent of theneuroendocrinereproductiveaxis,theyareinturn asthefinalgatekeepers Although GNRHneuronsserve The GNRHneuronal network as influentialthoseofleptin,insulinandghrelin. conveying nutritionalstatuscentrallydonotappeartobe as wellglucose( ( some nutritional signals, including insulin and ghrelin reproductive axis. 2014 it appears information regarding peripheral energy status processers andintegratorsof individualnutritionalsignals, fail-safe. InsteadofGNRHneurons actingasthereceivers, metabolic controloffertility tobemorefine-tunedand seem redundant and inefficient, itprobably allowsthe reproduction largelyoccursupstreamofGNRHneurons. the neuroendocrineintegrationofnutritionalsignalson ( hypothalamic circuitry afferenttoGNRHneurons its neuroendocrineeffectsappeartobemediatedthrough in brainslicepreparations( although it can modulate GNRH neurons directly, at least role inobesity-relatedinfertility. With regardtoghrelin, suggesting directinsulin–GNRHsignalingmayplaya 2007 from high-fatdiet-inducedinfertility( Interestingly, however, GNRH-IRKOmicewereprotected 2009 IRKO mice) ( respectively, GNRH-specificdeletionofInsR(GNRH- of LepR( ( although miceexhibitingbrain-specificdeletionofInsR regulation offertilitybynutritionalfactors.Forexample, does notplayacriticalrolewhenitcomestothe in mostcases,directhormone–GNRHcommunication effects thanatthelevelofindividualafferentneurons, the GNRHneuronalnetworkwouldhavemoreprofound regulation attheleveloffinalcommonoutput it mightbespeculatedthatdisruptionofmetabolic of reproduction Nutritional regulation Farkas Forbes Bruning Published byBioscientifica Ltd. Indeed, GNRHneuronsthemselves Although thisindirectmechanismofactionmay ), appearstoconvergeontheneuroendocrine ) and adiponectin ( ) didnotresultinanyreproductiveperturbations. et al et al Quennell et al . 2013 . 2009 Divall . 2000 , , et al t al et Evans ) orforebrainneuron-specificdeletion Zhang Schaeffer

. 2009 . 2010 Downloaded fromBioscientifica.com at09/24/202102:55:05PM et al Klenke et al ) weresubfertileandinfertile, Farkas et al . 2014 ) or LepR ( . 2007 . 2013 t al et t al et a , ), PYY( 58 . 2014 DiVall DiVall can . 2013 ). It thus appears : 2 Comninos Quennell directlydetect et al Pinilla ). Although et al ), manyof . 2015 . 2015 R110 et al t al et t al et via freeaccess ), ), . . . Review m c evans and g m anderson Nutritional regulation 58:2 R111 of reproduction

is first channeled through upstream circuitry. Information concentration within the GNRH neuron (Herbison & encoded by individual nutritional signals thus becomes Moenter 2011). contextualized among the information encoded by the To investigate the role(s) of leptin and insulin others, yielding a comprehensive snapshot of peripheral signaling via GABAergic neurons in the control of energy status. The GNRH neurons then receive this HPG axis function, our lab generated mice exhibiting integrated information and can respond accordingly, GABA-specific deletion of LepR Zuure( et al. 2013) and thereby maintaining their role as the final gatekeepers of InsR (Evans et al. 2014b) and demonstrated that leptin reproductive function. signaling, but not insulin signaling, via GABAergic Characterizing the and/or neurons is a major component of the circuitry identities of this ‘fuel-gauging’ network whereby peripheral metabolic status is relayed to the subserving GNRH neurons, as well as their role in neuroendocrine reproductive and metabolic axes. Mice mediating the metabolic control of fertility, remains lacking LepR from GABA neurons were profoundly an active area of research. Although many questions obese and exhibited and subfertility remain unanswered, a lot of headway has been made (Zuure et al. 2013), whereas mice lacking InsR from (Table 1). Perhaps not surprisingly, many of the same GABA neurons exhibited a very mild metabolic neuronal populations previously identified to play a role and normal pubertal development and in energy balance homeostasis have now been identified fertility (Evans et al. 2014b). Although it remains a to play a role in modulating reproductive function as possibility that the reproductive impairments observed well. Conversely, many neuronal populations previously in the GABA-specific LepR-knockout mice were directly known to modulate GNRH release are now recognized as related to their obesity phenotype, our lab and others integrators of nutritional information. have demonstrated that reproductive competency can be maintained despite profound obesity (Bates et al. 2003, Singireddy et al. 2013), suggesting leptin–GABA GABA-expressing neurons signaling is directly involved in the control of HPG axis While emphasis is often placed on the role of neuropeptide- function. In contrast, insulin–GABA signaling does not expressing populations with regard to the integration of appear to be critically involved in regulating fertility. metabolic and reproductive status, the contribution of Although leptin signaling via GABAergic neurons also deserves attention. It has been appears to play a critical role in the regulation of fertility, known since the 1980s that gamma-amino butyric many GABAergic neurons co-express other transmitters

Journal of Molecular Endocrinology acid (GABA) (the main inhibitory neurotransmitter in (Table 2), including NPY and AgRP (Meister 2007), the mammalian central nervous system) is involved in (Cheong et al. 2015) and dopamine (Olson & the regulation of GNRH release both before and after Nestler 2007, Marshall et al. 2016), for example, and it is puberty (Adler & Crowley 1986, Nikolarakis et al. 1988, therefore possible that both the neuropeptide(s) and/or Donoso et al. 1994, Mitsushima et al. 1994), and that neurotransmitter(s) released from these neurons act as GABAergic afferents compose a major group of synaptic the effectors on GNRH neurons. At this stage, it is not inputs to GNRH neurons. Similarly, it has been known clear whether leptin signaling via GABAergic neurons for some time that GABAergic neurons are modulated critically regulates fertility by modulating GABA release by nutritional factors, including leptin (Ovesjo et al. or the release of other transmitters, or both. However, 2001). However, it was not until Sullivan and coworkers evidence for the latter is strong. Furthermore, although demonstrated that GABAergic neurons play a role in insulin signaling via GABAergic neurons themselves conveying metabolic information to GNRH neurons does not appear to play a critical role (Evans et al. (Sullivan et al. 2003, Sullivan & Moenter 2004) that 2014b), insulin was shown to increase the number of

the role of GABAergic neurons in the integration of synaptic GABAA receptors to postsynaptic domains, nutritional and reproductive status became appreciated. which is the limiting factor in eliciting a greater

Indeed, the GNRH neurons express both GABAA and synaptic response (Wan et al. 1997, Bell-Horner et al.

GABAB receptor isoforms. Binding of GABA to the GABAB 2006). Therefore, insulin actions on GABAergic efferent receptor causes an inhibition of pulsatile GNRH release, neurons, via modulation of the GABA postsynaptic

but the effect of GABAA receptor activation can be either response, may explain some of insulin’s central effects excitatory or inhibitory depending on the chloride on GNRH release.

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology

Leptin Genetic background Table 1

Insulin DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Agrp nNOS Kiss1 Camk2a Gnrh Pomc Sf1 Nestin neo/neo +PMV-AAV-Flp LepR POMC Agrp db/db ob/ob ob/ob ob/ob ob/ob ob/ob ob/ob Vglut2 Vgat Gnrh Gnrh Kiss1 Pomc Agrp Vglut2 Vgat Review -cre + -cre + -cre + -cre + -cre + -cre + -cre + -Cre + -cre + -cre + -cre + -cre + -cre +

-cre + + + -cre + -cre + -cre + -cre + Nos1 Nos1 Gene deletionstudies:summaryofthemetaboliccontrolfertility. -cre + LepR LepR InsR LepR InsR LepR LepR InsR InsR InsR LepR LepR InsR LepR LepR InsR LepR InsR −/− −/− -flox LepR -flox -flox -flox -flox -flox -flox -flox -flox -flox -flox -flox -stop-flox -flox -flox -flox -stop-flox -flox -flox Genetic phenotype AgRP-specific Nitric oxide Kisspeptin- Forebrain neuron- GNRH-specific POMC-specific SF-1-specific LepR Brain-specific InsR PMV-restricted POMC-restricted Agrp-restricted LepR-deficient Leptin-deficient Leptin-deficient Leptin-deficient Leptin-deficient Leptin-deficient Leptin-deficient Glutamate- GABA-specific GNRH-specific GNRH-specific Kisspeptin- POMC-specific AgRP-specific Glutamate- GABA-specific LepR KO neurons specific LepRKO specific LepRKO LepR KO LepR KO KO KO LepR expression LepR expression LepR expression specific LepRKO LepR KO InsR KO InsR KO specific InsRKO InsR KO LepR KO specific InsRKO InsR KO m

c © 2017SocietyforEndocrinology

evans and Printed inGreatBritain g

m

anderson Manipulation – – – – – – – – – – – – – – Leptin-treated – Agrp-deficient Agrp-ablated Leptin-treated – HFD feeding – – – – – – of reproduction Nutritional regulation Published byBioscientifica Ltd. phenotype Metabolic Overweight Obese Normal body Obese Normal body Overweight Overweight Overweight Obese Mildly overweight Obese Overweight Overweight Obese Restored body Improved body Obese Restored body Restored body Obese Obese Normal body Normal body Normal Normal body Normal body Mildly overweight weight weight weight control weight control weight control weight control weight weight weight weight phenotype Reproductive Normal Impaired estrus Normal Infertile Normal Normal Normal Normal Delayed puberty, Normal Rescued fertility – Rescued fertility Infertile Infertile Infertile Rescued fertility Rescued fertility Rescued fertility Infertile Protected from Normal Normal Normal Normal Normal Normal

fertility cyclicity, normal subfertile infertility HFD-induced Downloaded fromBioscientifica.com at09/24/202102:55:05PM 58 References van deWall Leshan Donato Quennell Quennell van deWall Bingham Zuure Zuure Bruning Donato Huo O Egan&GM Hummel Bellefontaine Bellefontaine Sheffer-Babila Wu Mounzih Ingalls DiVall Divall Evans Konner Konner Evans Evans (2009) (2009) (2010) (2008) (2008) lished observations) Anderson (unpub (2014) (2014) (2013) Zhang : 2 et al et al et al et al et al et al et al et al et al et al , et al

et al et al et al et al et al . (2012) et al Shi et al . (2009) (Continued) et al et al et al et al . (2010) . (2014 . (2014 . (2014 . (2013) . (2013) . (2015) . (1950)

. (2012) et al et al et al . (2007) . (2007) . (2011) . (2011) . (1994) . (2000) . (1966) R112 . (1997) . (2008) et al et al . . et al . . . a b b . . ) ) ) - , via freeaccess . Review m c evans and g m anderson Nutritional regulation 58:2 R113 of reproduction

Table 1 Continued.

Metabolic Reproductive Genetic background Genetic phenotype Manipulation phenotype phenotype References Camk2a-cre + InsR-flox Forebrain – Mildly overweight Normal Evans et al. (2015) neuron-specific InsR KO Leptin + insulin Pomc-cre + InsR-flox + LepR-flox POMC-specific – Overweight Subfertile Hill et al. (2010) LepR and InsR KO Kiss1-cre + InsR-flox + LepR-flox Kisspeptin- – Normal body Altered pubertal Qiu et al. (2015) specific LepR weight timing and InsR KO Other Ghrelin−/− Ghrelin-deficient – Normal body Normal Sun et al. (2003) weight Ghsr−/− Ghrelin receptor- – Underweight – Sun et al. (2008) deficient Nestin-Cre + Sf1 flox/− Brain-specific SF1 – Normal body Impaired estrus Kim et al. (2010) KO weight cyclicity, subfertile

Glutamate-expressing neurons LH secretion occurs in a kisspeptin-independent manner (Garcia-Galiano et al. 2012). Furthermore, neurons in the Glutamate is the primary excitatory neurotransmitter ventral premamillary nucleus (PMV) of the hypothalamus, used by the majority of neurons in the CNS, and a body which mediate some of leptin’s metabolic influences of evidence demonstrates glutamate regulates GNRH on GNRH function (Donato et al. 2011), are almost neurons’ excitability and thus secretion. As reviewed exclusively glutamatergic (Kocsis et al. 2003), supporting by Iremonger and coworkers (Iremonger et al. 2010), a role for glutamate-expressing neurons in the metabolic many studies have shown that glutamate and control of fertility. antagonists injected into the brain can stimulate or To investigate the direct effects of leptin and insulin inhibit LH secretion, respectively (Lopez et al. 1990, Brann signaling via glutamatergic neurons in the metabolic & Mahesh 1995, Ping et al. 1997). Glutamate likely exerts regulation of fertility, our lab generated glutamate-

Journal of Molecular Endocrinology its influence via direct effects on GNRH neurons, as Tena- specific LepR KO and InsR KO mice. In contrast to the Sempere’s lab demonstrated that glutamate-stimulated

Table 2 Neuropeptide and transmitter co-expression.

Primary neuropeptide/ transmitter population Sub-population Co-expression Estimated co-expression (%) References AgRP/NPY ARC GABA ≤100 Marshall et al. (2016) POMC/CART ARC GABA <40 Wittmann et al. (2013) ARC Glutamate <60 Wittmann et al. (2013) Kisspeptin AVPV Glutamate <25 Cheong et al. (2015) AVPV Dopamine ≤50 Skrapits et al. (2015) ARC GABA <25 Cheong et al. (2015) ARC Glutamate <75 Cheong et al. (2015) ARC GABA <15 Marshall et al. (2016) nNOS ARC GABA <50 Marshall et al. (2016) SF1 VMH Glutamate ≤75 Tong et al. (2007a,b) VMH GABA ≤20 Tong et al. (2007a,b) Dopamine ARC GABA ≤80 Marshall et al. (2016) VTA GABA ≤40 Merrill et al. (2015) VTA Glutamate ≤20 Kawano et al. (2006) AVPV Kisspeptin ≤50 Skrapits et al. (2015)

AgRP, agouti-related peptide; ARC, ; AVPV, antero-ventral peri-ventricular nucleus; CART, cocaine-amphetamine-related-transcript; nNOS, neuronal nitric oxide synthase; NPY, ; POMC, pro-opiomelanocortin; SF1, steroidogenic factor 1; VMH, ventro-medial hypothalamus; VTA, ventral tegmental area.

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology the fasting-inducedsuppression ofLHreleasethatwas under normalconditions, they wereprotectedagainst knockout micedidnotexhibit afertilityphenotype ( expression andconcomitant suppressionofGNRHdrive ( reproductive function. are thus aptly equipped to integrate metabolic and GNRH release( thereby promotingNPY/AgRP-inducedsuppression of leptin andinsulin,ghrelinstimulatesNPY/AgRPneurons, ( (MC4) signaling stimulatory mediated, atleastinpart,viaAgRP’s antagonismof effectsofAgRPonGNRHneuronsarelikely inhibitory AgRP onGNRHneurons.Itshouldbenotedthatthe 2007 AgRP-expressing neurons( ( byghrelin-expressingneurons and arealsoinnervated insulin ( NPY/AgRP neurons,inturn,aredirectlymodulatedby Cone 2005 effectsonGNRHneurons( inhibitory orexigenic neuropeptidesshowntohavepredominantly (NPY) andagouti-relatedpeptide(AgRP),whichareboth (ARC) ofthehypothalamusco-expressneuropeptideY A subsetofGABAergicneurons in thearcuate nucleus NPY/AgRP-expressing neurons critically involved. but unlikeleptinsignalinginGABAneurons,itisnot to permit puberty onset and reproductive function, leptin signaling in glutamate neurons may be sufficient at theonsetofpuberty. Althoughspeculative,itappears suggest thePMVisakeysiteforleptin’s permissiveaction PMV is almost exclusively glutamatergic, and these data fertility ( LepR showedrescuedpubertyonsetandimproved exhibiting PMV-restricted re-expressionofendogenous Forexample,LepR-nullmice evidence suggestsotherwise. mediator ofthemetaboliccontrolfertility, some Although thesestudiessuggestglutamateisnotacritical exhibited normalHPGaxisfunction( ( LepR KOmiceexhibited normal pubertyonsetandfertility puberty andreducedfecundity, theglutamate-specific GABA-specific LepRKOmice,whichexhibiteddelayed Ollmann Cowley Zuure Kalamatianos Korner DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review Physiologically, fastingcausesanincrease in ), thereby reducing the inhibitory toneofNPY/ ), therebyreducingtheinhibitory et al t al et Konner Donato t al et et al , . 2013 Vulliemoz . 2001 . 2003 t al et . 1997 Lebrethon et al et al ). Glutamate-specificInsRKOmicealso ) and . 2008 . 2011 . 2007 ). InsulinandleptininhibitNPY/ , et al Cowley et al Npy ). Interestingly, although NPY- ). Asmentionedpreviously, the Sato . 2005 ) andleptin( ( et al . 2007 et al Brady m ,

Roa &Herbison2012 c . 1999 © 2017SocietyforEndocrinology

evans ). NPY/AgRPneurons . 2005 McShane t al et Evans and Elias ). Incontrast to Printed inGreatBritain . 1990 , Konner et al g et al

et al m

anderson . 2014 ) mRNA . 1999 . 1992 et al Agrp b ). ). ) . ,

and peptide,despitebeingenergyreplete,insulin show increasedhypothalamicexpressionof activation. Accordingly, insulin-deficientdiabeticrats ghrelin receptorpromotestonicNPY/AgRPneuronal Schwartz 2004 in responseto a fast ( observed may furtherpromotetheincreasedNPYexpression an increaseincirculating ghrelinconcentration,which 1997 fasted animals( in wild-type control mice ( observed ‘rescue’ mice,theNPY/AgRP-LepR ‘rescue’miceexhibited onset ( exclusively inthePMVwasalso sufficienttoinducepuberty sufficient topermitfertility ( that leptin signalingexclusivelyin NPY/AgRP neurons is neurons exert on reproductive function and also reveal tone NPY/AgRP-expressing both the powerful inhibitory involved inthesuppressionofGNRH.Thesedatahighlight respectively), it appears that they are both independently response tonutritionalstress(fastingandLepRdeficiency, shown toexhibitimprovedreproductivephenotypes in both NPY-knockout miceandAgRP-deficientwere AgRP-induced inhibitionofreproductivefunction.As NPY/AgRP signalingalonecansufficientlyblockNPY/ demonstratingthatleptin- unpublished observations), in NPY/AgRPneurons(OEgan&GMAnderson, deficient mice by restoring leptin sensitivity exclusively LepR- fertility canbecompletelyreinstatedinotherwise alone (vsneuronalablation)inLepR-null( tolerance ( restore their fertility, food intake regulation and glucose in leptin-deficient( was shownthatablatingNPY/AgRP-expressingneurons must restrainNPY/AgRPneuronstopermitfertility, it NPY/AgRP activity. suggests insulin is critically involved in restraining tonic administration restoredNPYlevels( have constitutiveactivity( However, asmentionedpreviously, ghrelinreceptors or leptindecreased thought tomediatethiseffect,astreatmentwithinsulin inresponsetofastingare insulin concentrationsobserved to lowenergyavailability. Thedecreasedleptinand whereby HPGfunctionbecomessuppressedinresponse suggesting NPYplaysacriticalroleinthemechanism Babila was alsoabletorescuetheirHPGaxisfunction( of reproduction Nutritional regulation Published byBioscientifica Ltd. As mentionedpreviously, reactivationofLepR Further supportingtheideathatinsulinand/orleptin , t al et Korner Donato Wu . 2013 et al et al et al ), so even in the absence of fasting,the Schwartz ). Lastly, ourlabrecentlyshowedthat . 2001 . 2011 . 2012 Npy

ob/ob mRNAandpeptideexpressionin ). Fasting is also accompanied by Downloaded fromBioscientifica.com at09/24/202102:55:05PM ). However, unlikethePMV-LepR et al ). Additionally, AgRPdeficiency ) diabeticmicewasableto Vulliemoz Holst . 1991 Asakawa t al et Abe Hill & Levine 2003 , 58 Wang &Leibowitz et al et al . 2003 : 2 t al et . 2005 . 1991 db/db Npy , Holst & . 2001 ). Sheffer- mRNA R114 ) mice ). This via freeaccess ). ). ), Review m c evans and g m anderson Nutritional regulation 58:2 R115 of reproduction

a slightly attenuated body weight phenotype. Although it POMC neurons, such that in the absence of leptin-POMC thus remains a possibility that the improved reproductive signaling, sufficient compensation occurs via insulin, and function observed in the NPY/AgRP-LepR ‘rescue’ mice vice versa. However, despite exhibiting subfertility, the might be due in part to their improved body weight, this mice lacking both leptin- and insulin-POMC signaling seems unlikely as, in addition to the PMV-LepR ‘rescue’ were still able to reproduce, suggesting insulin and leptin experiment, there are several accounts demonstrating can act via other circuitry to permit sufficient activation that fertility can be maintained in morbidly obese mice of the HPG axis, further highlighting how redundancy in (Bates et al. 2003, Singireddy et al. 2013). the mechanisms whereby nutritional signals reach GNRH neurons confers reproductive resilience.

POMC/CART-expressing neurons Kisspeptin-expressing neurons Acting in opposition to the NPY/AgRP neurons are ARC neurons encoding pro-opiomelanocortin (POMC, a The endogenous G-protein-coupled receptor 54 (GPR54) precursor of the anorexigenic melanocortin peptides) , kisspeptin, has been identified as the most potent and cocaine–amphetamine-related transcript (CART) secretagogue of GNRH (Irwig et al. 2004) and is likewise (POMC/CART neurons). These neurons play a key role in implicated in the timing of puberty onset (Han et al. energy homeostasis by promoting satiety and suppressing 2005) and the mechanism whereby energy status is feeding (Baskin et al. 1999, Meister 2007); yet, they have relayed to the reproductive axis (Tena-Sempere 2006, also been shown to promote GNRH neuron activity, Castellano et al. 2010, De Bond et al. 2016). Although presumably via direct actions (Leranth et al. 1988, Roa kisspeptin gene (Kiss1) expression is primarily regulated & Herbison 2012, True et al. 2013). Sub-populations of by gonadal steroid hormones (Smith 2009), it is also POMC neurons have been shown to co-express GABA modulated by metabolic status such that its expression and glutamate (Table 2). Perhaps not surprisingly, POMC/ becomes reduced in response to metabolic stress, such CART neurons are regulated by insulin, leptin and ghrelin as fasting (Castellano et al. 2005) or prolonged high-fat (Havel et al. 2000, Cowley et al. 2001, 2003, Balthasar et al. diet feeding (Quennell et al. 2011). Approximately 40% of 2004, Williams et al. 2010). Peripheral ghrelin peptide ARC kisspeptin neurons were shown to express the gene indirectly inhibits the activity of POMC/CART neurons for LepR (Smith et al. 2006) in one report, but subsequent in mice (Cowley et al. 2003), but POMC/CART neurons work suggests a more modest level of co-expression may also be directly modulated by ghrelin-expressing (Louis et al. 2011, Cravo et al. 2013) and failed to show

Journal of Molecular Endocrinology neurons via synaptic communication (Guan et al. 2008). direct leptin–kisspeptin signaling (Quennell et al. 2011). Leptin activates POMC neurons to promote satiety, and InsR protein expression was also observed in a subset of in the absence of leptin-POMC signaling, an obesity kisspeptin immunoreactive neurons (Evans et al. 2014a). phenotype results (Balthasar et al. 2004). Insulin’s Physiologically, kisspeptin treatment to women with appetite-suppressing effects are also largely mediated via acquired GNRH deficiency due to low energy availability POMC neurons (Benoit et al. 2002); yet, in the absence of was able to stimulate GNRH/LH release (Jayasena et al. direct insulin-POMC signaling, mice exhibit normal body 2009, 2010), suggesting that neuroendocrine integration weight regulation (Konner et al. 2007), presumably due of nutritional signals converge upstream of, or directly to the developmental compensation provided by leptin. on, kisspeptin neurons. Accordingly, leptin treatment to These data suggest leptin actions on POMC neurons are ob/ob mice increased Kiss1 mRNA expression (Smith et al. required for normal energy homeostasis, whereas insulin 2006), whereas ghrelin administration was shown to actions are not. reduce kisspeptin expression and LH pulse frequency With regard to fertility, insulin or leptin actions alone in ad libitum fed rats (Forbes et al. 2009). Also, insulin- on POMC neurons do not appear to play a critical role in dependent diabetic rats exhibit reduced kisspeptin mRNA conveying energy status to the HPG axis, as mice exhibiting levels (Castellano et al. 2006). Interestingly, insulin POMC-specific deletion of either LepR Balthasar ( et al. administration did not rescue the reduced kisspeptin 2004) or InsR (Konner et al. 2007) exhibited completely observed in these diabetic rats; yet, leptin normal fertility. Interestingly, mice exhibiting POMC- administration was sufficient to restoreKiss1 mRNA levels specific deletion of both LepR and InsR did present with and LH concentration (Castellano et al. 2006). reduced fertility, but not infertility (Hill et al. 2010). This Although ample evidence suggests leptin and insulin suggests leptin and insulin exert overlapping control of modulate kisspeptin neurons, their direct influences are

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology sufficient torescuetheonset ofpubertyinfood-restricted ( which occursinakisspeptin-independent manner to stimulateLHreleasein mice ( status totheHPGaxis. equipped totransduceinformationregardingnutritional by insulin( Cunningham regulated by leptin ( neurons. In turn, GALP neurons express LepR and are 2006 with enhancedgreenfluorescentprotein( in transgenicratswhichGNRHneuronsweretagged and axo-dendriticsynapticcontactswithGNRHneurons terminalswerealsofoundtomakeaxo-somatic nerve the rathypothalamus( fibers makeapparent contacts withGNRH neurons in cell bodies are primarily located in the ARC, but their Crown to themetaboliccontroloffertility( represent anotherpopulationofneuronsthatcontributes -like peptide(GALP)-expressingneurons GALP-expressing neurons cause reproductiveimpairments( to adulthoodenergyhomeostasis( of themelanocortinsystemandthuscausedisturbances which areknowntoimpairhypothalamicdevelopment explain, atleastinpart,whyprenatalnutritionalinsults, Pomc ARC kisspeptinneuronswererecentlyshowntoarisefrom downstream ofPOMCneurons.Interestingly, overhalfof conveyed toGNRHneuronscouldbebothupstreamand role inthemechanismwherebynutritionalstatusis activity ( POMC neuronsandindirectlyinhibitNPYneuronal 2016 indirectly viaPOMCneurons( likely thatleptin’s effectsonkisspeptinaremediated kisspeptin–GNRH signalingpathway, anditisthus recently documentedanovelleptin–melanocortin– ( fertile, yet,haddelayedpubertaltiming completely specific deletionofbothInsRandLepRwerealso normal HPG axis function. Mice exhibiting kisspeptin- ( exhibiting kisspeptin-specific deletion ofeither InsR not requiredfornormalreproductivecontrol,asmice Garcia-Galiano Qiu Evans DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review Physiologically, GALP administration was shown et al ), suggestingGALPcandirectlyregulateGNRH ). Conversely, kisspeptinwasshowntodirectlyexcite -expressing progenitors( et al t al et . 2015 Fu &vandenPol2010 . 2007 . 2014 Fraley et al ). However, Manfredi-Lozanoandcoworkers t al et , . 2002 a t al et Shioda ) orLepR( Jureus . 2012 Takatsu . 2004 ) andarealsodirectlymodulated et al et al ), andGALPinfusionwas . 2011 Sanz et al ). GALPneuronsarethus Donato . 2000 ), soitappears kisspeptin’s m Dupont Manfredi-Lozano

Kauffman Wattez et al c . 2001 © 2017SocietyforEndocrinology ). Anatomically, GALP

evans , Cunningham 2004 Takatsu t al et . 2015 and et al ). GALP-positive et al Printed inGreatBritain Takenoya . 2011 t al et g . 2012 . 2013 ). Thismay

et al m

anderson . 2005 ) retain . 2001 t al et ), can ). et al ), . , . , SF1 peptidedeletion.Inamore refinedexperiment,itwas phenotypewasadirectresultof whether theobserved and neuronalorganization, soitremainsunknown mice also exhibited impaired hypothalamic development obesity ( expression leads to impaired fertility and late-onset 2010 ( key roleinglucosehomeostasisandappetitecontrol hypothalamus (VMH),whichisknowntoplay a restricted to,andlargelycomprises,theventro-medial factor 1 (SF1) ( co-express ( Like GABAergicneurons,manyglutamatergicneurons SF1-expressing neurons coordinate theireffectsonfeedingbehaviorandfertility. neurons mayexhibitbi-directionalcommunicationto actions ( were neurons inthedorsomedialnucleusofhypothalamus their activity( on fertility, asGALPwasnotabletodirectlymodulate AgRP neuronsareinvolvedinmediatingGALP’s effects However, itdoesnotappearARCPOMC/CARTorNPY/ was abletodirectlystimulateGNRHneuronsisolated ( and insulin to rescue reproductive function in these rats demonstrating thatGALPmediatestheabilityofleptin was preventedwhenaGALPantibodyadministered, function in insulin-dependent diabetic rats; yet, this effect and leptinco-administrationrescuedreproductive of GALPexpression.Furthermore,subcutaneousinsulin suggesting insulinandleptinexhibitoverlappingcontrol Galp 48-h fast,andinsulinorleptinadministrationincreased Galp exhibiting insulin-dependentdiabeteshavereduced and alsofacilitatesfertility demonstrating GALPismodulatedbynutritionalsignals weanling maleandfemalerats( Kiss1 via kisspeptinneurons,asGALPadministrationincreased neurons. However, GALP’s effectsmayalsobemediated previously, GNRH GALPneuronsdirectlyinnervate from therat( administration reversedthiseffect( mRNA comparedtocontrolanimals,andbraininsulin of reproduction Nutritional regulation Tong Stoyanovitch Published byBioscientifica Ltd. GALP’s effectsonHPGfunctionmaybedirect,asGALP showntomediateGALP’s acuteorexigenicfeeding mRNAlevels( mRNAlevelswerealsoreducedinratsexposedtoa ). Interestingly, brain-specific deletion of SF1 peptide mRNAinfood-restrictedrats( t al et Kuramochi Kim . 2007 et al Kuramochi t al et Kuramochi Tong b . 2005 . 2010 ), aswellreproduction( et al Jureus

t al et . 2006 Downloaded fromBioscientifica.com at09/24/202102:55:05PM ). et al ). However, theseSF1-deficient Table 2 et al t al et . 2007 ), suggestingGALPandNPY . 2005 in vivo . 2000 . 2005 ), such as steroidogenic a ). SF1 expression is ) and,asmentioned Mohr , Fraley . Accordingly, rats ). Incontrast,NPY 58 Mohr Fraley : 2 t al et t al et t al et et al Kim . 2012 . 2004 . 2012 . 2004 R116 t al et Galp via freeaccess ). ), ). ), .

Review m c evans and g m anderson Nutritional regulation 58:2 R117 of reproduction

demonstrated that SF1 neuron-specific deletion of LepR neurotransmitters contribute to the regulation of hedonic leads to obesity without causing reproductive impairments eating. Nevertheless, these studies identify a role for both (Bingham et al. 2008). The former finding, albeit with its the ARC and VTA TH-expressing neurons in the regulation considerable caveats, may suggest SF1 peptide is critically of feeding behavior. involved in the regulation of fertility; yet, the latter With regard to fertility, dopamine has been shown to result suggests that leptin signaling via SF1-expressing exert both acute and tonic inhibition of GNRH neuron neurons is not, despite its critical role in regulating energy activity (Liu & Herbison 2013). Using electrophysiology homeostasis. Leptin’s roles in the control of metabolism in mouse brain slices, Liu and Herbison demonstrated and reproduction therefore appear dissociated, further that bath application of dopamine potently inhibited the highlighting the complexity of the neuroendocrine firing rate of ~50% of GNRH neurons, whereas application integration of metabolic and reproductive function. of dopamine receptor antagonists increased the basal firing of ~1/3 of GNRH neurons. Approximately 20% of GNRH neurons were shown to receive dopaminergic innervation Tyrosine hydroxylase-expressing neurons from the anteroventral (Liu & Tyrosine hydroxylase (TH)-expressing neurons, Herbison 2013); however, identifying whether the ARC which synthesize and release catecholamines (either or VTA dopamine population(s) are involved in the noradrenaline or dopamine), are also modulated by leptin, regulation of GNRH remains unclear. insulin and ghrelin (Hommel et al. 2006, Konner et al. In order to characterize the functional role of insulin 2011, Zhang & van den Pol 2016) and are involved and leptin signaling via dopamine neurons, mice exhibiting in the regulation of energy homeostasis (Khanh et al. TH-specific deletion of InsR or dopamine-specific deletion 2014) and fertility (Clarkson & Herbison 2011). Several of LepR were generated. Interestingly, although the distinct TH-expressing neuronal populations have been TH-specific InsR KO mice exhibited an increased body identified and anatomically characterized; yet, the major weight phenotype (Konner et al. 2011), the dopamine- populations thought to be involved in the regulation of specific LepR KO mice showed completely normal feeding energy homeostasis are the ARC and ventral tegmental behavior and body weight, even when challenged with area (VTA) dopaminergic populations. The TH neurons a high-fat diet (Liu et al. 2011). Unfortunately, their in the ARC, referred to as tuberoinfundibular dopamine reproductive phenotypes were not assessed. (TIDA) neurons, were recently shown to play an orexigenic role in homeostatic feeding behavior (Zhang & van den NOS-expressing neurons Journal of Molecular Endocrinology Pol 2016). Accordingly, ghrelin was shown to evoke direct excitatory effects in these neurons. A subset of these Nitric oxide synthase (NOS) neurons, which produce neurons were also shown to co-express GABA, which nitric oxide, comprise a subset of hypothalamic neurons was involved in mediating some of the orexigenic effects recently shown to mediate leptin’s effects on the HPG (Zhang & van den Pol 2016). The TH-expressing neurons axis. As mentioned previously, leptin treatment restores in the VTA of the midbrain are thought to be involved HPG axis function in leptin-deficient mice; yet, in the in the regulation of hedonic (vs homeostatic) feeding absence of nitric oxide signaling, leptin was unable behavior (Volkow et al. 2011). Accordingly, intra-VTA to induce puberty in ob/ob mice (Bellefontaine et al. leptin infusion was shown to decrease dopamine neuron 2014). This suggests nitric oxide signaling facilitates firing and reduce feeding Trinko( et al. 2011), whereas leptin’s permissive effects on puberty onset and ghrelin infusion into the VTA stimulated dopamine release fertility. Furthermore, neuronal NOS-knockout mice are and dramatically increased food intake (Abizaid 2009). hypogonadic and infertile (Gyurko et al. 2002), and mice Although insulin does not appear to directly modulate exhibiting NOS neuron-specific deletion of LepR exhibit dopamine release, it was shown to increase dopamine delayed puberty and hyperphagic obesity (Leshan et al. reuptake and thus, like leptin, caused an overall reduction 2012). However, even though NOS expression identified in dopamine signaling, leading to suppressed food intake the population of neurons involved in mediating leptin’s (Mebel et al. 2012). Although dopamine appears to be the effects on metabolic and reproductive function, the major effector of these observations, sub-populations of authors suggest it is unlikely that nitric oxide signaling VTA dopamine neurons have been shown to co-express itself was responsible for leptin’s observed metabolic GABA (Olson & Nestler 2007) and glutamate (Alsio et al. effects because NOS-deficient mice show no obvious 2011), and it thus remains a possibility that these metabolic phenotype (Huang 2000). Sub-populations of

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology disruption of neural signal transducer and activator regard tothemetaboliccontrol offertility. Forexample, for discretesignalingpathways havebeenestablishedwith 2011 Carvalheira Schwartz 2003 demonstrated ( to theirrespective receptors being activated has been between theintracellularpathwaysactivatedinresponse for example,yetahighdegreeofoverlapandinteraction exert theirinfluenceonspecificneuronalpopulations, Leptin andinsulinmightactviauniquereceptors to intracellular signalingmoleculesshouldnotbeignored. of metabolic and reproductive function, the role of hormone–neuron interactionsinvolvedintheintegration emphasis isoftenplacedonidentifyingthespecific Intracellular signalingmolecules Other considerations ultimately regulateGNRHdrive. inputs from the GNRH neuronal network that stimulatory and still contributetothenetimpactofallinhibitory ignored as under normal physiological conditions, they of these‘lesserinvolved’populationsshouldnotbe involved ( of reproductivefunctionappearstobemorecritically 2015 2007 energy availabilitysuppressesGNRHdrive( are alsoimplicatedinthemechanismwherebylow signaling ( regulation ofRFRPneuronsappearstooccurviaindirect LepR ( hormone neurons,wererecentlyshowntoco-express mammalian orthologstoaviangonadotropin-inhibiting small subpopulationofRFRPneurons,whicharethe hormone (MCH)andorexin-expressingneurons.A RFamide-related peptide(RFRP),melanin-concentrating involvement ofothers.Amongthese,forexample,are control offertility, there isalsoevidence supporting the arguably thekeypopulationsinvolvedinmetabolic Although theaforementionedneuronalpopulationsare Other neuronal populations downstream metaboliceffects. one oftheseneurotransmittersisresponsibleforleptin’s glutamate ( NOS neurons co-express GABA ( DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review , ), yet,theinfluenceofNPY/AgRPinsuppression , Burgos-Ramos Backholer Poling Rizwan Wu Lin et al et al , et al Niswender t al et Mirshamsi et al . 2005 et al . 2014 . 2004 . 2012 et al . 2009 , . 2014 Morris &Rui2009 . 2011 ), forexample.However, leptin’s ), anditisthereforepossiblethat et al et al ). Nevertheless,theinfluence , Wu ). MCH and neurons ). Furthermore,criticalroles . 2004 . 2001 et al Marshall m

c © 2017SocietyforEndocrinology

evans , . 2009 , Benomar 2003 and et al Printed inGreatBritain , , , Berthou Skrapits Niswender & g Chen Although . 2016

et al m

anderson . 2005 t al et et al et al ) and . , . . reproductive function remained intact ( STAT3 inactivationwasrestrictedtoLepR-expressingcells, inactivation ofleptin-inducedsignaling.However, when in theseSTAT3-knockout micemightbeduetothe activation, onemightassumethephenotypeobserved infertility inmice( of transcription3(STAT3) causesobesity, diabetesand mTOR signaling,phosphatidylinositol-3-kinase(PI3K) Roa &Tena-Sempere 2014 puberty infood-restrictedfemalerats( puberty andalsobluntedtheabilityofleptintopromote that mTORinactivationinhibitedthegonadotropicaxisat reproductive function.Roaandcoworkersdemonstrated has beenidentifiedasacentralregulatorofmetabolicand to amultitudeoffactors,includinginsulinandleptin, of rapamycin(mTOR), which canbeactivatedinresponse reproductive function.Similarly, themammaliantarget leptin-induced STAT3 activation may be required for 2013 release ( the effectsoffastingtosuppressGNRH-stimulatedLH Moenter (2011) availability playsarole.Asreviewedby yet, thereisalsoampleevidencesuggestingglucose play an important role in the metabolic control of fertility; intracellular signalingcascadestheyinitiateundoubtedly Cellular energysensing nutritional signalsonfertility. molecules playintheneuroendocrineintegrationof understanding oftherolesthatintracellularsignaling others, demonstratetheimportanceofamorecomplete by reproductive functionbecomessuppressed,asreviewed are thought to play a role in the mechanism whereby chronic under-andover-nutrition,theseimpairments in response to insulin-induced PI3K signaling are observed and reproductivefunction.Impairmentsinleptin-and/or signaling isalsoconsideredakeyintegratorofmetabolic hypothalamic glucosesensing, whichinturncould there isevidencesuggesting nutritionalsignalsmodulate induced LHsecretion( override thefasting-induced suppressionofGNRH- energy balance(i.e.fasted or fed),yet,wasunableto led toanincreaseinLHconcentrationregardless of For example,blockadeofK perturbations viatheseK K experiments showGNRHneuronsexpressATP-sensitive of reproduction Nutritional regulation + Published byBioscientifica Ltd. (K Acosta-Martinez (2011) ), highlighting that STAT3 signaling other than ATP Bucholtz ) channelsandaresensitivetometabolic , adecreaseinglucoseavailabilitymediates t al et Gao

et al Huang . 1996 Downloaded fromBioscientifica.com at09/24/202102:55:05PM ATP . 2004 ATP channels( ). InadditiontoSTAT3 and

. Thesedata,alongwith channelswithtolbutamide Nutritional signalsandthe , t al et Howland 1980 ). AsleptininducesSTAT3 . 2008 58 Zhang : Roa 2 Singireddy ). Furthermore, t al et Roland and et al ). . 2007 . 2009 In vivo R118 t al et via freeaccess ). . ,

Review m c evans and g m anderson Nutritional regulation 58:2 R119 of reproduction

influence GNRH neuronal function (Bruning et al. neuron adhesiveness in the mouse hypothalamus 2000, Diggs-Andrews et al. 2010). Insulin and leptin, (Sandau et al. 2011b), and it was shown that astrocyte-to-

for example, have been shown to activate KATP channels GNRH neuron adhesiveness via SynCAM1 is important in (Spanswick et al. 2000, Mirshamsi et al. 2004). the control of female sexual development. For example, monophosphate (AMP)-activated protein female mice expressing a SynCAM1 dominant negative kinase (AMPK), long considered a cell-autonomous form specifically in astrocytes exhibited delayed onset of energy sensor, can also act as an integrative metabolic puberty, disrupted estrus cyclicity and reduced fecundity sensor in the brain (Ramamurthy & Ronnett 2006) and (Sandau et al. 2011a). Astroglial cells, therefore, play a may therefore modulate GNRH neuronal activity, either role in GNRH neuronal function and may facilitate the directly or indirectly by stimulating or inhibiting neurons nutritional regulation of fertility. in the GNRH neuronal network. The activity of AMPK Tanycytes, which are specialized bipolar glial cells is normally regulated by immediate glucose availability, located in the ARC and median eminence, also play a but is also increased in the presence of orexigenic role in GNRH release via extension and retraction of their stimuli (e.g. AgRP and ghrelin) and decreased in the end-foot processes between GNRH synaptic terminals presence of anorexigenic stimuli (e.g. insulin and leptin) (Rodriguez et al. 2005, Baroncini et al. 2007). As reviewed (Minokoshi et al. 2004). This permits the possibility that by Levin and coworkers, tanycytes express tight junctions whole body energy status (i.e. fuel reserves) acts in concert that regulate the permeability of the brain parenchyma with immediate glucose availability to modulate the to CSF and may therefore have the capacity to integrate activity of neurons. Minokoshi and coworkers propose a number of signals associated with energy status a model for how orexigenic and anorexigenic signals, (Levin et al. 2004). Tanycytes express glucose transporters,

through AMPK activation and suppression, respectively, as well as KATP channels (Garcia et al. 2003), which supports result in an integrated metabolic response required the possibility that they act as glucose sensors and as a for the regulation of food intake and energy balance possible conduit whereby peripheral nutritional signals (Minokoshi et al. 2004), and this could presumably be the could modulate GNRH neuronal activity. Accordingly, case for reproductive function as well. tanycytes were shown to respond rapidly to changes in glucose and ATP concentration (Frayling et al. 2011). Astroglial cells It is also possible that astroglial cells are involved in the mechanism whereby nutritional signals Stress responses as metabolic modulators of HPG modulate GNRH neuronal activity (Baroncini et al. 2007, axis function Although the evidence supporting the Journal of Molecular Endocrinology Ojeda et al. 2008). Firstly, astrocytes play an important linkage between metabolic impairments and reproductive role in glucose metabolism (Yi et al. 2011), which, as disturbances is strong, others have proposed the idea of a discussed previously, is an important regulator of GNRH stress-related mechanism in which reproductive function neuronal function. Secondly, leptin and insulin signaling is compromised secondary to the activation of the stress- via astrocytes has been demonstrated (Yi et al. 2011). response systems: the hypothalamic–pituitary–adrenal Astrocytic insulin signaling modulates glucose metabolism (HPA) and sympathoadrenal axes (Tilbrook et al. 2000, in primary human astrocytes (Heni et al. 2011) as well 2002). Although this review has focused on metabolic as in vivo (Garcia-Caceres et al. 2016), which suggests stress, it has also been shown that immune stress (i.e. insulin actions on astrocytes could indirectly mediate inflammation), psychosocial stress, environmental stress the effects of glucose availability on GNRH neuronal and physical stress are associated with alterations in GNRH function. Leptin signaling in astrocytes was also recently secretion and subsequent reproductive disturbances shown to play an active role in the hypothalamic control (O’Byrne et al. 1988, Chen et al. 1992, Li et al. 2004a,b, of feeding (Kim et al. 2014). Recently, astrocytes were Berga 2006, 2008, Kinsey-Jones et al. 2009, Knox et al. shown to control food intake by inhibiting AgRP neuron 2009). It therefore stands to reason that stress itself activity (Yang et al. 2015), which suggests astrocytes exerts an inhibitory effect on reproductive function. are indirectly able to promote fertility as restraining However, some evidence suggests the influence of stress AgRP neurons would presumably facilitate GNRH drive, cannot be easily separated from its impact on nutritional as mentioned previously. Furthermore, Sandau and signals, suggesting the final common pathway leading to coworkers demonstrated that the synaptic cell adhesion compromised reproductive function may be perceived low molecule, SynCAM1, mediates astrocyte-to-GNRH energy availability (Loucks & Redman 2004, Berga 2008).

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology insulin andghrelinontheGNRHneuronalnetworkor primarily viathedirectsignalingofhormoneslikeleptin, of reproductive function due to metabolic stress occurs what remainstobedeterminediswhethersuppression of reproductivefunction( was showntoexertasynergisticeffectonthesuppression (that ontheirownhadlittleimpactHPGfunction) impact ofminormetabolicandpsychosocialstressors and stressfactors.Insupportofthis,thecombined is suppressed might be a convergence of both metabolic that thefinalpathwaywherebyreproductivefunction ( hypothalamus andbyblockadeofHPA axisactivation adrenergic signalingintheparaventricularnucleusof amplitude inratswaspreventedbytheblockadeof fasting-induced suppressionofLHpulsefrequencyand caused bytheactivationofstressaxes.Forexample, energy availability. induced metabolicprofileissimilartothatofchroniclow words, stresshasametabolicinfluence,andthestress- hypo-metabolic state( adaptive metabolic changes that are characteristic of a hormonesthatalterfuelregulationandsignal regulatory the bloodstream( and secretionofglucocorticoidscatecholaminesinto body’s stress-responsesystemsstimulatesthesynthesis behindthishypothesisisthatactivationofthe The theory Maeda DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review Conversely, however, nutritional infertility couldbe t al et . 1994 Tilbrook ). Itthereforeremainsapossibility Laughlin &Yen 1996 Williams et al . 2000 m

et al c © 2017SocietyforEndocrinology

evans ), whicharecounter- . 2007 and Printed inGreatBritain g ). Therefore,

). Inother m

anderson ( signaling consistentlycausesreproductiveimpairments pathway. However, widespreaddeletionofcentralleptin the truephysiologicalimportanceoftargeted signals and/or neuronal mediators) and often undermines compensation viaotherpathways(e.g.nutritional reproductive function( mediator doesnotappeartojeopardizeneuroendocrine individual metabolichormonesthroughanyoneneuronal the metaboliccontroloffertility, impairedsignalingof Due tothecomplexityandinherentredundancyof Summary responsesisalsorequired. secretory whether activationofthesympathoadrenalandHPA and AgRPdeficiency( the demonstrationthatAgRPablation( competency, respectively. Thislatterfinding,alongwith mice wassufficienttoreinstatepubertyandreproductive inLepR-null GM Anderson,unpublishedobservations) the deletion ofLepR signaling from discrete neuronal normal reproductivecontrol.Furthermore,although 2016 mice alsorestorefertility, highlightsthatunrestrained PMV ( axis function,restoringLepRsignalingexclusivelyinthe populations consistentlyfailedtomarkedlyimpairHPG of reproduction Nutritional regulation Mounzih Published byBioscientifica Ltd. ), highlightingleptinisabsolutelyrequiredfor Donato t al et et al . 1997 . 2011 area. *,co-expressiondataisnotavailable. steroidogenic factor-1; VTA, ventraltegmental amphetamine-regulated transcript; SF1, POMC/CART, pro-opiomelanocortin/cocaine- and kisspeptin; nNOS,neuronalnitricoxide synthase; DA, dopamine;GALP, galanin-like peptide;Kiss, AgRP/NPY, agouti-relatedpeptide/neuropeptide Y; receptor donotdisruptreproductivedrive). not shownasthedeletionofghrelinorits not criticallyinvolved(e.g.effects ofghrelinare effects. Pathwaysnotrepresentedbyarrowsare arrows indicatecombinedleptinandinsulin orange arrowsindicateinsulineffects andgreen literature. Bluearrowsindicateleptineffects, potency oftheeffect basedonthereviewed thickness ofarrowrepresentsanestimatethe Cre-LoxP genedeletionexperiments).The involved arrows representpathwaysshowntobecritically absolutely requiredforfertility, whereasdotted Solid arrowsrepresentpathwaysidentifiedtobe circles) theneuroendocrinereproductiveaxis. signals topromote(greycircles)orsuppress(black neuronal populationstargetedbynutritional Schematic diagramillustratingthemajor Figure 1

, Sheffer-Babila Quennell ) or in AgRP neurons (O Egan and Downloaded fromBioscientifica.com at09/24/202102:55:05PM i. 1 Fig.

but not required (as identified through but notrequired(asidentifiedthrough ). Thisislikelydueto t al et et al . 2009 58 : 2 Wu . 2013 , t al et Zuure ) in . 2012 R120 t al et ob/ob via freeaccess ) .

Review m c evans and g m anderson Nutritional regulation 58:2 R121 of reproduction

AgRP expression is critically involved in the suppression diabetic rats. Brain Research 539 223–227. (doi:10.1016/0006- of HPG function due to nutritional stress. 8993(91)91624-A) Abizaid A 2009 Ghrelin and dopamine: new insights on the peripheral In contrast, widespread deletion of central insulin regulation of appetite. Journal of Neuroendocrinology 21 787–793. signaling (Bruning et al. 2000, Evans et al. 2014b, 2015) (doi:10.1111/j.1365-2826.2009.01896.x) or ghrelin signaling (Sun et al. 2008) does not cause Acosta-Martinez M 2011 PI3K: an attractive candidate for the central integration of metabolism and reproduction. Frontiers in infertility, demonstrating they are not required to permit Endocrinology 2 110. (doi:10.3389/fendo.2011.00110) sufficient HPG activation. This is not to say they are Adler BA & Crowley WR 1986 Evidence for gamma-aminobutyric acid not important modulators of GNRH drive, but rather modulation of ovarian hormonal effects on secretion and hypothalamic catecholamine activity in the female rat. highlights sufficient compensation can occur in their Endocrinology 118 91–97. (doi:10.1210/endo-118-1-91) absence to preserve reproductive function. However, Ahima RS, Prabakaran D, Mantzoros C, Qu D, Lowell B, Maratos-Flier E pathological insulin signaling via GNRH neurons due & Flier JS 1996 Role of leptin in the neuroendocrine response to fasting. Nature 382 250–252. (doi:10.1038/382250a0) to diet-induced obesity promotes infertility (DiVall et al. Alsio J, Nordenankar K, Arvidsson E, Birgner C, Mahmoudi S, Halbout B, 2015), demonstrating insulin can directly influence HPG Smith C, Fortin GM, Olson L, Descarries L, et al. 2011 Enhanced axis function. Furthermore, it remains a possibility that sucrose and cocaine self-administration and cue-induced drug seeking after loss of VGLUT2 in midbrain dopamine neurons in insulin critically regulates reproductive function through mice. Journal of Neuroscience 31 12593–12603. (doi:10.1523/ its combined actions on the hypothalamus and pituitary JNEUROSCI.2397-11.2011) (Brothers et al. 2010). Asakawa A, Inui A, Kaga T, Yuzuriha H, Nagata T, Ueno N, Makino S, Fujimiya M, Niijima A, Fujino MA, et al. 2001 Ghrelin is an appetite- In conclusion, the neuroendocrine integration stimulatory signal from stomach with structural resemblance to of metabolic and reproductive function is complex, . Gastroenterology 120 337–345. (doi:10.1053/gast.2001.22158) and many nutritional signals play a role in conveying Backholer K, Smith J & Clarke IJ 2009 Melanocortins may stimulate reproduction by activating orexin neurons in the dorsomedial information regarding peripheral energy status centrally hypothalamus and kisspeptin neurons in the of the to the GNRH neuronal network. Due to the inherent ewe. Endocrinology 150 5488–5497. (doi:10.1210/en.2009-0604) redundancies in the metabolic control of fertility, the Balthasar N, Coppari R, McMinn J, Liu SM, Lee CE, Tang V, Kenny CD, McGovern RA, Chua SC Jr, Elmquist JK, et al. 2004 use of transgenic models to test the functional role of signaling in POMC neurons is required for normal body weight individual metabolically relevant signaling pathways homeostasis. Neuron 42 983–991. (doi:10.1016/j.neuron.2004.06.004) has largely been unsuccessful. Nevertheless, these Barash IA, Cheung CC, Weigle DS, Ren H, Kabigting EB, Kuijper JL, Clifton DK & Steiner RA 1996 Leptin is a metabolic signal to the investigations have importantly demonstrated how reproductive system. Endocrinology 137 3144–3147. (doi:10.1210/ adaptable and robust the neuroendocrine reproductive en.137.7.3144) axis is in regard to its ability to integrate and respond to Baroncini M, Allet C, Leroy D, Beauvillain JC, Francke JP & Prevot V 2007 Morphological evidence for direct interaction between Journal of Molecular Endocrinology multiple metabolic cues. gonadotrophin-releasing hormone neurones and astroglial cells in the human hypothalamus. Journal of Neuroendocrinology 19 691–702. (doi:10.1111/j.1365-2826.2007.01576.x) Baskin DG, Figlewicz Lattemann D, Seeley RJ, Woods SC, Porte D Jr & Declaration of interest Schwartz MW 1999 Insulin and leptin: dual adiposity signals to the The authors declare that there is no conflict of interest that could be brain for the regulation of food intake and body weight. Brain perceived as prejudicing the impartiality of this review. Research 848 114–123. (doi:10.1016/S0006-8993(99)01974-5) Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, et al. 2003 STAT3 signalling is required for leptin regulation of energy balance but not Funding reproduction. Nature 421 856–859. (doi:10.1038/nature01388) M C E was supported by The Health Sciences Career Development Program Beak SA, Heath MM, Small CJ, Morgan DG, Ghatei MA, Taylor AD, Postdoctoral Fellowship from the University of Otago, and research was Buckingham JC, Bloom SR & Smith DM 1998 Glucagon-like supported by the Health Research Council of New Zealand. peptide-1 stimulates luteinizing hormone-releasing hormone secretion in a rodent hypothalamic neuronal cell line. Journal of Clinical Investigation 101 1334–1341. (doi:10.1172/JCI610) Belgardt BF & Bruning JC 2010 CNS leptin and insulin action in the Author contribution statement control of energy homeostasis. Annals of the New York Academy of M C E drafted the manuscript and G M A provided support and edited the Sciences 1212 97–113. (doi:10.1111/j.1749-6632.2010.05799.x) final manuscript. Bell-Horner CL, Dohi A, Nguyen Q, Dillon GH & Singh M 2006 ERK/ MAPK pathway regulates GABAA receptors. Journal of Neurobiology 66 1467–1474. (doi:10.1002/neu.20327) References Bellefontaine N, Chachlaki K, Parkash J, Vanacker C, Colledge W, de Tassigny XD, Garthwaite J, Bouret SG & Prevot V 2014 Leptin- Abe M, Saito M, Ikeda H & Shimazu T 1991 Increased neuropeptide Y dependent neuronal NO signaling in the preoptic hypothalamus content in the arcuato-paraventricular hypothalamic neuronal facilitates reproduction. Journal of Clinical Investigation 124 system in both insulin-dependent and non-insulin-dependent 2550–2559. (doi:10.1172/JCI65928)

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology Carvalheira JB,TorsoniCarvalheira MA,UenoM,Amaral ME,AraujoEP, Velloso LA, Carro E,PinillaL,SeoaneLM,ConsidineRV, AguilarE,CasanuevaFF& Cameron JL&NosbischC1991Suppressionofpulsatileluteinizing- Burgos-Ramos E,ChowenJA,Arilla-FerreiroCanellesS,ArgenteJ& Bucholtz DC,Vidwans NM,HerbosaCG,SchilloKK&FosterDL1996 Bruning JC,GautamD,BurksDJ,GilletteJ,SchubertM,OrbanPC, Brothers KJ,Wu S,DiVall SA,MessmerMR,KahnCR,MillerRS, Brann DW&MaheshVB1995Glutamate:amajorneuroendocrine Brady LS,SmithMA,GoldPW&HerkenhamM1990Alteredexpression Boden G,ChenX,MozzoliM&RyanI1996Effectoffastingonserum Bingham NC,AndersonKK,ReuterAL,StallingsNR&ParkerKL2008 Berthou F, RouchC,GertlerA,GerozissisK&Taouis M2011Chronic Berga SL,MortolaJF, GirtonL,SuhB,LaughlinG,PhamP&Yen SS Berga SL2008Stressandreproduction:ataleoffalsedichotomy? Berga S2006Stress,metabolismandreproductivecompromise. Benomar Y, Wetzler S,Larue-AchagiotisC,DjianeJ,Tome D&Taouis M Benoit SC,AirEL,CoolenLM,StraussR,JackmanA,CleggDJ,SeeleyRJ DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review 48–57. leptin signalingsystemsinrathypothalamus. Gontijo JA&SaadMJ2005Cross-talk betweentheinsulinand Neuroendocrinology cycle andluteinizinghormonepulsatility infemalerats. Dieguez C1997Influenceofendogenousleptintoneontheestrous Endocrinology restriction intheadultmalerhesus-monkey(macaca-mulatta). hormone andtestosteronesecretionduringshort-termfood (doi:10.1111/j.1471-4159.2011.07191.x) ofNeurochemistry Journal insulin receptorwithIRS2andincreasingitsassociationSOCS3. to acutecentralinsulininjectionbyreducingtheinteractionof Barrios V2011Chroniccentralleptininfusionmodifiestheresponse Endocrinology luteinizing hormonesecretionisdependentonglucoseavailability. Metabolic interfacesbetweengrowthandreproduction.V. Pulsatile 289 insulin receptorincontrolofbodyweightandreproduction. Klein R,KroneW, Muller-Wieland D&KahnCR2000Roleofbrain (doi:10.1016/j.cmet.2010.06.010) knockout oftheinsulinreceptor. induced infertilityinfemalemiceduetoapituitary-specific Radovick S,Wondisford FE&Wolfe A2010Rescueofobesity- 325–329. secretion. signalmediatingsteroideffectsongonadotropin excitatory (doi:10.1159/000125626) deprived rats. of hypothalamicneuropeptidemRNAsinfood-restrictedandfood- Metabolism leptin innormalhumansubjects. Endocrinology nucleus resultsinincreasedadiposityandametabolicsyndrome. Selective lossofleptinreceptorsintheventromedialhypothalamic (doi:10.1016/j.mce.2011.02.005) Metabolism Endocrinology a002529) liver andhypothalamus. 2005 Invivoleptininfusionimpairsinsulinandsignallingin mediated bymelanocortins. & Woods SC2002Thecatabolicactionofinsulininthebrainis signaling. central leptininfusiondifferentlymodulatesbrainandliverinsulin hypothalamic amenorrhea. 1989 Neuroendocrineaberrationsinwomenwithfunctional Reproduction 59–66. 2122–2125. (doi:10.1038/oby.2005.7) (doi:10.1016/j.mce.2005.07.003) (doi:10.1016/0960-0760(95)00070-G) Journal of Steroid Biochemistry andMolecularBiology ofSteroid Biochemistry Journal Molecular andCellularEndocrinology

81 68

21

128 137 149 149 3419–3423. 301–308. Neuroendocrinology I32–I32. (doi:10.1126/science.289.5487.2122) 1532–1540. 601–607. 2138–2148. 867–868.

66 375–377.

(doi:10.1210/jcem-68-2-301) (doi:10.1093/oxfordjournals.humrep. 117 Molecular andCellularEndocrinology (doi:10.1210/jc.81.9.3419) Journal ofClinicalEndocrinologyand Journal (doi:10.1210/en.137.2.601) (doi:10.1210/en.2008-0004) Journal ofNeuroscience Journal 175–185. (doi:10.1210/endo-128-3-1532) (doi:10.1210/en.2007-1200) (doi:10.1159/000127262)

52 Cell Metabolism Journal ofClinicalEndocrinologyand Journal 441–447. m

c © 2017SocietyforEndocrinology

evans

337 Obesity Research and

12 Printed inGreatBritain 89–95.

22 295–305. g 9048–9052.

m

anderson

Human

242 53 Science

13

Cone RD2005Anatomyandregulationofthecentralmelanocortin Compagnucci C,GE,LomnicziA,MohnVacas I, Cowley MA,PronchukN,FanW, DinulescuDM, ColmersWF& Corr M,DeSouzaMJ,Toombs RJ&Williams NI2011Circulating leptin Comninos AN,JayasenaCN&DhilloWS2014Therelationship Codner E,MerinoPM&Tena-Sempere M2012Femalereproductionand Clarkson J&HerbisonAE2011Dualphenotypekisspeptin-dopamine and Christian CA&MoenterSM2010Theneurobiologyofpreovulatory Cheong RY, CzieselskyK,PorteousR&HerbisonAE2015Expressionof Chen LL,JiangQY, ZhuXT, ShuG,BinYF, Wang XQ,GaoP&Zhang YL Chen MD,O’ByrneKT, ChiappiniSE,HotchkissJ&KnobilE1992 Chehab FF, MounzihK,LuR&LimME1997Earlyonsetof Chan JL,HeistK,DePaoliAM,Veldhuis JD&MantzorosCS2003The Castellano JM,BentsenAH,MikkelsenJD&Tena-Sempere M2010 Castellano JM,NavarroVM,Fernandez-FernandezR,RoaJ,Vigo E, Castellano JM,NavarroVM,Fernandez-FernandezR,NogueirasTovar S, of reproduction Nutritional regulation Published byBioscientifica Ltd. 6273(00)80829-6) basis fortheadipostat. in thehypothalamicparaventricular nucleus:evidenceofacellular 1999IntegrationofNPY,Cone RD AGRP, andmelanocortinsignals deq375) women. concentrations donotdistinguishmenstrualstatusinexercising system. (doi:10.1159/000067177) the growingmalerat. axisin of nutritionalstressonthehypothalamo-pituitary-gonadal Cebral E,ElverdinJC,FriedmanS,RettoriV&BoyerPM2002Effect Reproduction Update between gutandadiposehormones,reproduction. Reproduction Update type 1diabetes:frommechanismstoclinicalfindings. (doi:10.1111/j.1365-2826.2011.02107.x) Neuroendocrinology project togonadotrophin-releasinghormoneneurones. neurones oftherostralperiventricularareathirdventricle Neuroendocrinology generator activityintherhesusmonkey:roleofovary. Hypoglycemic ‘stress’andgonadotropin-releasinghormonepulse Science reproductive functioninnormalfemalemicetreatedwithleptin. Clinical Investigation inhealthymen. adaptation toshort-termstarvation role offallingleptinlevelsintheneuroendocrineandmetabolic Research Reviews -induced gonadotropin-releasinghormonesurges. JNEUROSCI.1776-15.2015) ofNeuroscience Journal puberty onset,estrogenfeedback,andfertilityinfemalemice. ESR1 inglutamatergicandGABAergicneuronsisessentialfornormal 893–902. AMolecularandIntegrativePhysiology Physiology: Part on ontogenyandfeedingcondition. proventriculus andliverofchicken(Gallusgallusdomesticus):studies 2007 Ghrelinligand-receptormRNAexpressioninhypothalamus, Kisspeptins: bridgingenergyhomeostasisandreproduction. 1584) diabetic malerats. gonadotropic responsesbykisspeptininstreptozotocin-induced Expression ofhypothalamicKiSS-1systemandrescuedefective Pineda R,DieguezC,AguilarE,PinillaL&Tena-Sempere M2006 Endocrinology activation ofthereproductiveaxisbykisspeptininundernutrition. Changes inhypothalamicKiSS-1systemandrestorationofpubertal Roa J,Vazquez MJ,Vigo E,CasanuevaFF, AguilarE,

275 31

Nature Neuroscience 1364 Human Reproduction (doi:10.1016/j.cbpa.2007.02.017) 544–577. 88–90.

146 129–138. 3917–3925.

56 23

(doi:10.1126/science.275.5296.88) Diabetes

20 18 111 (doi:10.1210/er.2009-0023) 666–673. 293–301.

Neuroimmunomodulation 35 153–174. 568–585.

Neuron (doi:10.1016/j.brainres.2010.08.057) 1409–1421. 14533–14543.

8

Downloaded fromBioscientifica.com at09/24/202102:55:05PM

55 571–578. 26 (doi:10.1210/en.2005-0337)

24 2602–2610. 685–694. (doi:10.1159/000126291) (doi:10.1093/humupd/dmt033) (doi:10.1093/humupd/dms024) 155–163. Comparative Biochemistry and and Comparative Biochemistry (doi:10.1172/JCI200317490) (doi:10.1038/nn1455) (doi:10.1523/ (doi:10.1093/humrep/ (doi:10.1016/S0896- (doi:10.2337/db05- 58

10 : 2 153–162. et al

147 Journal of Journal Human . 2005 . 2005 Human Journal of Journal

Endocrine Brain R122 via freeaccess Review m c evans and g m anderson Nutritional regulation 58:2 R123 of reproduction

Cowley MA, Smart JL, Rubinstein M, Cerdan MG, Diano S, Horvath TL, POMC neurons projecting to the lateral hypothalamic area. Neuron Cone RD & Low MJ 2001 Leptin activates anorexigenic POMC 23 775–786. (doi:10.1016/S0896-6273(01)80035-0) neurons through a neural network in the arcuate nucleus. Nature Evans MC, Rizwan M, Mayer C, Boehm U & Anderson GM 2014a 411 480–484. (doi:10.1038/35078085) Evidence that insulin signalling in gonadotrophin-releasing Cowley MA, Smith RG, Diano S, Tschop M, Pronchuk N, Grove KL, hormone and kisspeptin neurones does not play an essential role in Strasburger CJ, Bidlingmaier M, Esterman M, Heiman ML, et al. 2003 metabolic regulation of fertility in mice. Journal of Neuroendocrinology The distribution and mechanism of action of ghrelin in the CNS 26 468–479. (doi:10.1111/jne.12166) demonstrates a novel hypothalamic circuit regulating energy Evans MC, Rizwan MZ & Anderson GM 2014b Insulin action on GABA homeostasis. Neuron 37 649–661. (doi:10.1016/S0896- neurons is a critical regulator of energy balance but not fertility in 6273(03)00063-1) mice. Endocrinology 155 4368–4379. (doi:10.1210/en.2014-1412) Cravo RM, Frazao R, Perello M, Osborne-Lawrence S, Williams KW, Evans MC, Rizwan MZ & Anderson GM 2015 Insulin does not target Zigman JM, Vianna C & Elias CF 2013 Leptin signaling in kiss1 CamkIIalpha neurones to critically regulate the neuroendocrine neurons arises after pubertal development. PLoS ONE 8 e58698. reproductive axis in mice. Journal of Neuroendocrinology 27 899–910. (doi:10.1371/journal.pone.0058698) (doi:10.1111/jne.12330) Crown A, Clifton DK & Steiner RA 2007 Neuropeptide signaling in the Farkas I, Vastagh C, Sarvari M & Liposits Z 2013 Ghrelin decreases firing integration of metabolism and reproduction. Neuroendocrinology 86 activity of gonadotropin-releasing hormone (GnRH) neurons in an 175–182. (doi:10.1159/000109095) estrous cycle and endocannabinoid signaling dependent manner. Cunningham MJ 2004 Galanin-like peptide as a link between PLoS ONE 8 e78178. (doi:10.1371/journal.pone.0078178) metabolism and reproduction. Journal of Neuroendocrinology 16 Farooqi IS, Jebb SA, Langmack G, Lawrence E, Cheetham CH, 717–723. (doi:10.1111/j.1365-2826.2004.01221.x) Prentice AM, Hughes IA, McCamish MA & O’Rahilly S 1999 Effects Cunningham MJ, Scarlett JM & Steiner RA 2002 Cloning and of recombinant leptin therapy in a child with congenital leptin distribution of galanin-like peptide mRNA in the hypothalamus and deficiency. New England Journal of Medicine 341 879–884. pituitary of the macaque. Endocrinology 143 755–763. (doi:10.1210/ (doi:10.1056/NEJM199909163411204) endo.143.3.8661) Farooqi IS, Matarese G, Lord GM, Keogh JM, Lawrence E, Agwu C, De Bond JP, Tolson KP, Nasamran C, Kauffman AS & Smith JT 2016 Sanna V, Jebb SA, Perna F, Fontana S, et al. 2002 Beneficial effects of Unaltered hypothalamic metabolic gene expression in Kiss1r KO leptin on obesity, T cell hyporesponsiveness, and neuroendocrine/ mice despite obesity and reduced energy expenditure. Journal of metabolic dysfunction of human congenital leptin deficiency. Journal Neuroendocrinology 28 jne.12430. (doi:10.1111/jne.12430) of Clinical Investigation 110 1093–1103. (doi:10.1172/JCI0215693) De Souza MJ, Leidy HJ, O’Donnell E, Lasley B & Williams NI 2004 Forbes S, Li XF, Kinsey-Jones J & O’Byrne K 2009 Effects of ghrelin on Fasting ghrelin levels in physically active women: relationship with Kisspeptin mRNA expression in the hypothalamic medial preoptic menstrual disturbances and metabolic hormones. Journal of Clinical area and pulsatile luteinising hormone secretion in the female rat. Endocrinology and Metabolism 89 3536–3542. (doi:10.1210/jc.2003- Neuroscience Letters 460 143–147. (doi:10.1016/j.neulet.2009.05.060) 032007) Fraley GS, Scarlett JM, Shimada I, Teklemichael DN, Acohido BV, Clifton De Souza MJ, Lee DK, VanHeest JL, Scheid JL, West SL & Williams NI DK & Steiner RA 2004 Effects of diabetes and insulin on the 2007 Severity of energy-related menstrual disturbances increases in expression of galanin-like peptide in the hypothalamus of the rat. proportion to indices of energy conservation in exercising women. Diabetes 53 1237–1242. (doi:10.2337/diabetes.53.5.1237) Fertility and Sterility 88 971–975. (doi:10.1016/j. Frayling C, Britton R & Dale N 2011 ATP-mediated glucosensing by fertnstert.2006.11.171) hypothalamic tanycytes. Journal of Physiology 589 2275–2286. Diggs-Andrews KA, Zhang X, Song Z, Daphna-Iken D, Routh VH & (doi:10.1113/jphysiol.2010.202051) Journal of Molecular Endocrinology Fisher SJ 2010 Brain insulin action regulates hypothalamic glucose Fu LY & van den Pol AN 2010 Kisspeptin directly excites anorexigenic sensing and the counterregulatory response to hypoglycemia. neurons but inhibits orexigenic neuropeptide Diabetes 59 2271–2280. (doi:10.2337/db10-0401) Y cells by an indirect synaptic mechanism. Journal of Neuroscience 30 Divall SA, Williams TR, Carver SE, Koch L, Bruning JC, Kahn CR, 10205–10219. (doi:10.1523/JNEUROSCI.2098-10.2010) Wondisford F, Radovick S & Wolfe A 2010 Divergent roles of growth Furuta M, Funabashi T & Kimura F 2001 Intracerebroventricular factors in the GnRH regulation of puberty in mice. Journal of Clinical administration of ghrelin rapidly suppresses pulsatile luteinizing Investigation 120 2900–2909. (doi:10.1172/JCI41069) hormone secretion in ovariectomized rats. Biochemical and DiVall SA, Herrera D, Sklar B, Wu S, Wondisford F, Radovick S & Wolfe A Biophysical Research Communications 288 780–785. (doi:10.1006/ 2015 signaling in the GnRH neuron plays a role in bbrc.2001.5854) the abnormal GnRH pulsatility of obese female mice. PLoS ONE 10 Gao Q, Wolfgang MJ, Neschen S, Morino K, Horvath TL, Shulman GI & e0119995. (doi:10.1371/journal.pone.0119995) Fu XY 2004 Disruption of neural signal transducer and activator of Donato J Jr, Cravo RM, Frazao R, Gautron L, Scott MM, Lachey J, transcription 3 causes obesity, diabetes, infertility, and thermal Castro IA, Margatho LO, Lee S, Lee C, et al. 2011 Leptin’s effect on dysregulation. PNAS 101 4661–4666. (doi:10.1073/pnas.0303992101) puberty in mice is relayed by the ventral premamillary nucleus and Garcia MA, Millan C, Balmaceda-Aguilera C, Castro T, Pastor P, does not require signaling in Kiss1 neurons. Journal of Clinical Montecinos H, Reinicke K, Zuniga F, Vera JC, Onate SA, et al. 2003 Investigation 121 355–368. (doi:10.1172/JCI45106) Hypothalamic ependymal-glial cells express the glucose transporter Donoso AO, Seltzer AM, Navarro CE, Cabrera RJ, Lopez FJ & Negro- GLUT2, a protein involved in glucose sensing. Journal of Vilar A 1994 Regulation of luteinizing hormone-releasing hormone Neurochemistry 86 709–724. (doi:10.1046/j.1471-4159.2003.01892.x) and luteinizing hormone secretion by hypothalamic amino acids. Garcia-Galiano D, van Ingen Schenau D, Leon S, Krajnc-Franken MA, Brazilian Journal of Medical and Biological Research 27 921–932. Manfredi-Lozano M, Romero-Ruiz A, Navarro VM, Gaytan F, van Dupont C, Cordier AG, Junien C, Mandon-Pepin B, Levy R & Chavatte- Noort PI, Pinilla L, et al. 2012 Kisspeptin signaling is indispensable Palmer P 2012 Maternal environment and the reproductive function for , but not glutamate, stimulation of gonadotropin of the offspring. Theriogenology 78 1405–1414. (doi:10.1016/j. secretion in mice. Endocrinology 153 316–328. (doi:10.1210/en.2011- theriogenology.2012.06.016) 1260) Elias CF, Aschkenasi C, Lee C, Kelly J, Ahima RS, Bjorbaek C, Flier JS, Garcia-Caceres C, Quarta C, Varela L, Gao Y, Gruber T, Legutko B, Saper CB & Elmquist JK 1999 Leptin differentially regulates NPY and Jastroch M, Johansson P, Ninkovic J, Yi CX, et al. 2016 Astrocytic

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology Huang W, Acosta-MartinezM,HortonTH&Levine JE2008Fasting- Huang PL2000Mousemodelsofnitricoxidesynthasedeficiency. Howland BE1980Effectofglucoprivationinducedby2-deoxy-d-glucose Hommel JD,Trinko R,SearsRM,GeorgescuD,LiuZW, GaoXB, Hill JW, EliasCF, Fukuda M,Williams KW, BerglundED,HollandWL, Hill JW&LevineJE2003Abnormalresponseoftheneuropeptide Herbison AE&MoenterSM2011Depolarisingandhyperpolarising Herbison AE2016Controlofpubertyonsetandfertilityby Holst B,CygankiewiczA,JensenTH,AnkersenM&SchwartzTW2003 Holst B&SchwartzTW2004Constitutiveghrelinreceptoractivityasa Heni M,HennigeAM,PeterA,Siegel-AxelD,OrdelheideKrebsN, Havel PJ,HahnTM,SindelarDK,BaskinDG,DallmanMF, Weigle DS& Han SK,GottschML,LeeKJ,PopaSM,SmithJT, JakawichSK, Gyurko R,LeupenS&HuangPL2002Deletionofexon6the Guan JL,OkudaH,Takenoya F, KintakaY, Yagi M,Wang L,SekiM, DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review ajpendo.90615.2008) Endocrinology andMetabolism ATP-sensitive potassiumchannels. induced suppressionofLHsecretion doesnotrequireactivationof oftheAmericanSocietyNephrology Journal (doi:10.1055/s-2007-999190) andMetabolicResearchHormone -induced releaseofluteinizinghormoneinrats. responsetoGnRH and on serumgonadotropinlevels,pituitary 51 signaling inmidbraindopamineneuronsregulatesfeeding. Thurmon JJ,MarinelliM&DiLeoneRJ2006Leptinreceptor (doi:10.1210/me.2003-0069) potent inverseagonist. High constitutivesignalingoftheghrelinreceptor–identificationa Sciences signaling set-pointinappetiteregulation. 286–297. normal glucosehomeostasisandfertility. leptin actiononpro-opiomelanocortinneuronsisrequiredfor Cho YR,ChuangJC,XuY, ChoiM, 221024) lactation. Y-deficient mousereproductiveaxistofooddeprivationbutnot (doi:10.1111/j.1365-2826.2011.02145.x) Neuroendocrinology hormone neurones:towardsanemergingconsensus. actions ofGABA(A)receptoractivationongonadotrophin-releasing Endocrinology gonadotropin-releasing hormoneneurons. astrocytes. human promotes glycogenstorageandcellproliferationinprimary Machicao F, FritscheA,HaringHU&StaigerH2011Insulin 244–252. muscle uncouplingprotein3expressioninrats. insulin treatmentonthehypothalamicmelanocortinsystemand Schwartz MW2000Effectsofstreptozotocin-induceddiabetesand Neuroscience neuroendocrine switchfortheonsetofpuberty. gonadotropin-releasing hormoneneuronsbykisspeptinasa SteinerRA&HerbisonAE2005Activationof Clifton DK, (doi:10.1210/endo.143.7.8921) hypogonadism andinfertility. neuronal nitricoxidesynthasegeneinmiceresults regpep.2007.09.028) arcuate nucleus. proopiomelanocortin- andghrelin-containingneuronsintherat Hori Y, KageyamaH&ShiodaS2008Synapticrelationshipsbetween availability. insulin signalingcouplesbrainglucoseuptakewithnutrient 801–810.

25 (doi:10.2337/diabetes.49.2.244) (doi:10.1016/j.cmet.2010.03.002) Endocrinology 113–117. PLoS ONE

Cell 25

(doi:10.1016/j.neuron.2006.08.023) 12 11349–11356.

452–466. 166 Regulatory Peptides Regulatory

23 867–880.

(doi:10.1016/j.tips.2004.01.010) 6 557–569. e21594.

Molecular Endocrinology 144 (doi:10.1038/nrendo.2016.70) 1780–1786.

295 (doi:10.1523/JNEUROSCI.3328-05.2005) Endocrinology (doi:10.1016/j.cell.2016.07.028)

12 (doi:10.1371/journal.pone.0021594) E1439–E1446. 520–523.

American Journal ofPhysiology: American Journal 145 et al m 128–132.

. 2010Directinsulinand (doi:10.1210/en.2002- c © 2017SocietyforEndocrinology

Cell Metabolism Trends inPharmacological evans

Nature Reviews

11 143 S120–S123.

17 and Journal of of Journal 2767–2774. Diabetes (doi:10.1152/ 2201–2210. Printed inGreatBritain (doi:10.1016/j. Journal of Journal g

m

49

11 anderson

Neuron

Jayasena CN,NijherGM,AbbaraA,MurphyKG,LimPatelD, Jayasena CN,NijherGM,ChaudhriOB,MurphyKG,RangerA,Lim Iremonger KJ,ConstantinS,LiuX&HerbisonAE2010Glutamate Ingalls AM,DickieMM&SnellGD1950Obese,anewmutationinthe Huo L,GamberK,GreeleyS,SilvaJ,HuntoonN,LengXH&BjorbaekC Hummel KP, DickieMM&ColemanDL1966Diabetes,anewmutation Kim JG,SuyamaS,KochM,JinArgente-Arizon P, ArgenteJ,Liu ZW, Kim KW, LiS, ZhaoH,PengB,Tobet SA,ElmquistJK,ParkerKL& Khanh DV, ChoiYH,MohSH,KinyuaAW &KimKW2014Leptinand Kawano M,KawasakiA,Sakata-HagaH,FukuiY, KawanoH,NogamiH Kauffman AS,BuenzleJ,FraleyGS&RissmanEF2005Effectsofgalanin- Katsiki N&HatzitoliosAI2010Insulin-sensitizingagentsinthe Kalra SP, UenoN&KalraPS2005Stimulationofappetitebyghrelinis Kalamatianos T, GrimshawSE,PoorunR,HahnJD&CoenCW2008 Jureus A,CunninghamMJ,McClainME,CliftonDK&SteinerRA2000 Irwig MS,FraleyGS,SmithJT,Irwig AcohidoBV, PopaSM,CunninghamMJ, of reproduction Nutritional regulation Published byBioscientifica Ltd. Mehta A, ToddMehta A, C,DonaldsonM,Trew GH, 0406) Endocrinology andMetabolism but chronicadministrationcausestachyphylaxis. gonadotropin secretioninwomenwithhypothalamicamenorrhea, Subcutaneous injectionofkisspeptin-54acutelystimulates Patel D,MehtaA,Todd C,RamachandranR, (doi:10.1159/000083140) mRNA inthemalerat. gonadotropin releasinghormoneneuronsandregulationofKiSS-1 Gottsch ML,CliftonDK&SteinerRA2004Kisspeptinactivationof (doi:10.1016/j.brainres.2010.08.071) regulation ofGnRHneuronexcitability. house mouse. activity byPOMCneurons. 2009 Leptin-dependentcontrolofglucosebalanceandlocomotor science.153.3740.1127) in themouse. Zimmer MR,JeongJK,Szigeti-Buck K, 1240–1250. impaired femalereproductivefunction. 2010CNS-specificablation ofsteroidogenicfactor1resultsin Zhao L (doi:10.3389/fpsyg.2014.00846) energy balanceandrewardsystem. insulin signalingindopaminergicneurons:relationshipbetween 581–592. transporter 2intheratbrain. hypothalamic dopamineneuronsexpressvesicularglutamate & HisanoS2006Particularsubpopulationsofmidbrainand (doi:10.1016/j.yhbeh.2005.01.010) in femaleandmalemice. like peptide(GALP)onlocomotion,reproduction,andbodyweight GCO.0b013e32833e1264) in ObstetricsandGynecology syndrome:anupdate. treatment ofpolycysticovary ofNutrition Journal regulated byleptinrestraint:peripheralandcentralsitesofaction. (doi:10.1111/j.1365-2826.2008.01757.x) ofNeuroendocrinology Journal neuropeptide YintheAVPV orarcuate nucleusoffemalerats. nucleus (AVPV): effectsoffastingontheexpressionKiSS-1and Fasting reducesKiSS-1expressionintheanteroventralperiventricular (doi:10.1210/en.141.7.2703) the hypothalamusofrat. Galanin-like peptide(GALP)isatargetforregulationbyleptinin clpt.2010.204) andTherapeutics Clinical Pharmacology reproductive hormonesinwomenwithhypothalamicamenorrhea. administration ofkisspeptin-54for8weeksstimulatesrelease (doi:10.1002/cne.21054) (doi:10.1210/me.2009-0206) Journal ofHeredity Journal Science

135

153

1331–1335. Neuroendocrinology 1127–1128. Hormones andBehavior Hormones

22 Downloaded fromBioscientifica.com at09/24/202102:55:05PM

Cell Metabolism

20 94 Endocrinology 466–476. Journal ofComparativeNeurology Journal 1089–1097.

4315–4323. 41 317–318. Frontiers inPsychology

88 et al (doi:10.1126/ Molecular Endocrinology Brain Research (doi:10.1097/ 840–847.

80 . 2014Leptinsignalingin

141 et al 9 58 (doi:10.1210/jc.2009- 264–272. 537–547. et al 2703–2706. : . 2010Twice-weekly 2

48 . 2009 Journal ofClinical Journal (doi:10.1038/ 141–151. Current Opinion

1364

5 846. 35–43. R124

498

24

via freeaccess Review m c evans and g m anderson Nutritional regulation 58:2 R125 of reproduction

astrocytes regulates hypothalamic neuronal circuits and feeding. Endocrinology and Metabolism 82 318–321. (doi:10.1210/ Nature Neuroscience 17 908–910. (doi:10.1038/nn.3725) jcem.82.1.3840) Kinsey-Jones JS, Li XF, Knox AM, Wilkinson ES, Zhu XL, Chaudhary AA, Laughlin GA, Morales AJ & Yen SS 1997 Serum leptin levels in women Milligan SR, Lightman SL & O’Byrne KT 2009 Down-regulation of with polycystic ovary syndrome: the role of insulin resistance/ hypothalamic kisspeptin and its receptor, Kiss1r, mRNA expression is hyperinsulinemia. Journal of Clinical Endocrinology and Metabolism 82 associated with stress-induced suppression of luteinising hormone 1692–1696. (doi:10.1210/jc.82.6.1692) secretion in the female rat. Journal of Neuroendocrinology 21 20–29. Laughlin GA, Dominguez CE & Yen SS 1998 Nutritional and endocrine- (doi:10.1111/j.1365-2826.2008.01807.x) metabolic aberrations in women with functional hypothalamic Klenke U, Taylor-Burds C & Wray S 2014 Metabolic influences on amenorrhea. Journal of Clinical Endocrinology and Metabolism 83 reproduction: adiponectin attenuates GnRH neuronal activity in female 25–32. (doi:10.1210/jc.83.1.25) mice. Endocrinology 155 1851–1863. (doi:10.1210/en.2013-1677) Lebrethon MC, Aganina A, Fournier M, Gerard A, Parent AS & Kluge M 2012 Ghrelin suppresses secretion of gonadotropins in women. Bourguignon JP 2007 Effects of in vivo and in vitro administration Reproductive Sciences 19 Np3–Np3. (doi:10.1177/1933719112459242) of ghrelin, leptin and neuropeptide mediators on pulsatile Kluge M, Schussler P, Uhr M, Yassouridis A & Steiger A 2007 Ghrelin gonadotrophin-releasing hormone secretion from male rat suppresses secretion of luteinizing hormone in humans. Journal of hypothalamus before and after puberty. Journal of Neuroendocrinology Clinical Endocrinology and Metabolism 92 3202–3205. (doi:10.1210/ 19 181–188. (doi:10.1111/j.1365-2826.2006.01518.x) jc.2007-0593) Leranth C, MacLusky NJ, Shanabrough M & Naftolin F 1988 Kluge M, Uhr M, Bleninger P, Yassouridis A & Steiger A 2009 Ghrelin Immunohistochemical evidence for synaptic connections between suppresses secretion of FSH in males. Clinical Endocrinology 70 pro-opiomelanocortin-immunoreactive axons and LH-RH neurons in 920–923. (doi:10.1111/j.1365-2265.2008.03440.x) the preoptic area of the rat. Brain Research 449 167–176. Kluge M, Schussler P, Schmidt D, Uhr M & Steiger A 2012 Ghrelin (doi:10.1016/0006-8993(88)91035-9) suppresses secretion of luteinizing hormone (LH) and follicle- Leshan RL, Greenwald-Yarnell M, Patterson CM, Gonzalez IE & stimulating hormone (FSH) in women. Journal of Clinical Endocrinology Myers MG 2012 Leptin action through hypothalamic nitric oxide and Metabolism 97 E448–E451. (doi:10.1210/jc.2011-2607) synthase-1-expressing neurons controls energy balance. Nature Kluge M, Schmidt D, Uhr M & Steiger A 2013 Ghrelin suppresses Medicine 18 820-U236. (doi:10.1038/nm.2724) nocturnal secretion of luteinizing hormone (LH) and thyroid Levin BE, Routh VH, Kang L, Sanders NM & Dunn-Meynell AA 2004 stimulating hormone (TSH) in patients with major depression. Neuronal glucosensing: what do we know after 50 years? Diabetes 53 Journal of Psychiatric Research 47 1236–1239. (doi:10.1016/j. 2521–2528. (doi:10.2337/diabetes.53.10.2521) jpsychires.2013.05.010) Li XF, Bowe JE, Mitchell JC, Brain SD, Lightman SL & O’Byrne KT 2004a Knox AM, Li XF, Kinsey-Jones JS, Wilkinson ES, Wu XQ, Cheng YS, Stress-induced suppression of the gonadotropin-releasing hormone Milligan SR, Lightman SL & O’Byrne KT 2009 Neonatal pulse generator in the female rat: a novel neural action for lipopolysaccharide exposure delays puberty and alters hypothalamic gene-related peptide. Endocrinology 145 1556–1563. Kiss1 and Kiss1r mRNA expression in the female rat. Journal of (doi:10.1210/en.2003-1609) Neuroendocrinology 21 683–689. (doi:10.1111/j.1365-­ Li XF, Edward J, Mitchell JC, Shao B, Bowes JE, Coen CW, Lightman SL 2826.2009.01885.x) & O’Byrne KT 2004b Differential effects of repeated restraint stress Kocsis K, Kiss J, Csaki A & Halasz B 2003 Location of putative on pulsatile lutenizing hormone secretion in female Fischer, Lewis glutamatergic neurons projecting to the medial preoptic area of the and Wistar rats. Journal of Neuroendocrinology 16 620–627. rat hypothalamus. Brain Research Bulletin 61 459–468. (doi:10.1016/ (doi:10.1111/j.1365-2826.2004.01209.x) S0361-9230(03)00180-1) Lin LH, Edwards RH, Fremeau RT, Fujiyama F, Kaneko T & Talman WT Journal of Molecular Endocrinology Konner AC, Janoschek R, Plum L, Jordan SD, Rother E, Ma X, Xu C, Enriori 2004 Localization of vesicular glutamate transporters and neuronal P, Hampel B, Barsh GS, et al. 2007 Insulin action in AgRP-expressing nitric oxide synthase in rat nucleus tractus solitarii. Neuroscience 123 neurons is required for suppression of hepatic glucose production. Cell 247–255. (doi:10.1016/j.neuroscience.2003.08.063) Metabolism 5 438–449. (doi:10.1016/j.cmet.2007.05.004) Liu X & Herbison AE 2013 Dopamine regulation of gonadotropin- Konner AC, Hess S, Tovar S, Mesaros A, Sanchez-Lasheras C, Evers N, releasing hormone neuron excitability in male and female mice. Verhagen LA, Bronneke HS, Kleinridders A, Hampel B, et al. 2011 Endocrinology 154 340–350. (doi:10.1210/en.2012-1602) Role for insulin signaling in catecholaminergic neurons in control of Liu J, Perez SM, Zhang W, Lodge DJ & Lu XY 2011 Selective deletion of energy homeostasis. Cell Metabolism 13 720–728. (doi:10.1016/j. the leptin receptor in dopamine neurons produces anxiogenic-like cmet.2011.03.021) behavior and increases dopaminergic activity in . Molecular Korner J, Savontaus E, Chua SC Jr, Leibel RL & Wardlaw SL 2001 Leptin Psychiatry 16 1024–1038. (doi:10.1038/mp.2011.36) regulation of Agrp and Npy mRNA in the rat hypothalamus. Journal Lopez FJ, Donoso AO & Negro-Vilar A 1990 Endogenous excitatory of Neuroendocrinology 13 959–966. neurotransmission regulates the estradiol-induced LH (doi:10.1046/j.1365-2826.2001.00716.x) surge in ovariectomized rats. Endocrinology 126 1771–1773. Kuramochi M, Kohno D, Onaka T, Kato S & Yada T 2005 Galanin-like (doi:10.1210/endo-126-3-1771) peptide and ghrelin increase cytosolic Ca2+ in neurons containing Loucks AB & Thuma JR 2003 Luteinizing hormone pulsatility is growth hormone-releasing hormone in the arcuate nucleus. disrupted at a threshold of energy availability in regularly Regulatory Peptides 126 85–89. (doi:10.1016/j.regpep.2004.08.017) menstruating women. Journal of Clinical Endocrinology and Metabolism Kuramochi M, Onaka T, Kohno D, Kato S & Yada T 2006 Galanin-like 88 297–311. (doi:10.1210/jc.2002-020369) peptide stimulates food intake via activation of neuropeptide Y Loucks AB & Redman LM 2004 The effect of stress on menstrual neurons in the hypothalamic dorsomedial nucleus of the rat. function. Trends in Endocrinology and Metabolism 15 466–471. Endocrinology 147 1744–1752. (doi:10.1210/en.2005-0907) (doi:10.1016/j.tem.2004.10.005) Laughlin GA & Yen SS 1996 Nutritional and endocrine-metabolic Loucks AB, Verdun M & Heath EM 1998 Low energy availability, not aberrations in amenorrheic athletes. Journal of Clinical Endocrinology stress of exercise, alters LH pulsatility in exercising women. Journal of and Metabolism 81 4301–4309. (doi:10.1210/jc.81.12.4301) Applied Physiology 84 37–46. Laughlin GA & Yen SS 1997 Hypoleptinemia in women athletes: absence Louis GW, Greenwald-Yarnell M, Phillips R, Coolen LM, Lehman MN & of a diurnal rhythm with amenorrhea. Journal of Clinical Myers MG Jr 2011 Molecular mapping of the neural pathways

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology Nandi A,Wang X, AcciliD&Wolgemuth DJ2010Theeffectofinsulin Mounzih K,LuR&ChehabFF1997Leptintreatmentrescuesthe Morris DL&RuiLY 2009Recentadvancesinunderstandingleptin Mohr MA,LeathleyE&FraleyGS2012Hypothalamicgalanin-like Mitsushima D,HeiDL&Terasawa E1994gamma-Aminobutyricacidis Mirshamsi S,LaidlawHA,NingK,AndersonE,BurgessLA,GrayA, Minokoshi Y, AlquierT, FurukawaN,KimYB,LeeA,XueB,MuJ, Merrill CB,FriendLN,NewtonST, HopkinsZH&EdwardsJG2015 Meister B2007Neurotransmittersinkeyneuronsofthehypothalamus Mebel DM,Wong JC,DongYJ&BorglandSL2012Insulin inthe McShane TM,MayT, MinerJL&KeislerDH1992Centralactionsof Marshall CJ,DesroziersE,McLennanT&CampbellRE2016Defining Manfredi-Lozano M,RoaJ,Ruiz-PinoF, PietR,Garcia-Galiano D, Maeda KI,CagampangFRA,CoenCW&Tsukamura H1994 MacLusky NJ,CookS,ScrocchiL,ShinJ,KimVaccarino F, AsaSL& DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review en.2009-0788) and hyperglycemia. signaling onfemalereproductivefunction independentofadiposity 1190–1193. sterility ofgeneticallyobeseob/obmales. ajpendo.00274.2009) Endocrinology andMetabolism signaling andleptinresistance. (doi:10.1111/j.1365-2826.2012.02351.x) weanling rats. peptide (GALP)rescuestheonsetofpubertyinfood-restricted 395–399. hormone-releasing hormonebeforetheonsetofpuberty. neurotransmitterrestrictingthereleaseof luteinizing an inhibitory 5 actin reorganizationandKATP channelactivation. signalling pathwaysinarcuate nucleusneurones:PI3Kdependent Sutherland C&AshfordML2004Leptinandinsulinstimulationof hypothalamus. food intakebyrespondingtohormonalandnutrientsignalsinthe Foufelle F, FerreP, BirnbaumMJ, srep16176) biosynthetic elements. properties andexpressionofmRNAforendocannabinoid Ventral tegmentalareadopamineandGABAneurons:Physiological Behavior that regulatefeedingbehaviorandbodyweight. Neuroscience dopamine concentrationviaincreasedreuptake. ventral tegmentalareareduceshedonicfeedingandsuppresses (doi:10.1095/biolreprod46.6.1151) nutrition andreproduction. linkbetween neuropeptide-Y mayprovideaneuromodulatory [in press]. and prenatallyandrogenizedfemalemice. subpopulations ofarcuate nucleusGABAneuronsinmale,female Metabolism pathway involvedinthemetaboliccontrolofpuberty. Ruiz A, ZamoraA,LeonS,Sanchez-GarridoMA,Romero- Pineda R, Endocrinology induced suppressionofluteinizing-hormonereleaseinfemalerats. nucleus andofcorticotropin-releasinghormoneinthefasting- Involvement ofthecatecholaminergicinputtoparaventricular en.141.2.752) receptor signaling. in micewithcompletedisruptionofglucagon-likepeptide-1 Drucker DJ2000Neuroendocrinefunctionandresponsetostress 152 linking leptintotheneuroendocrinereproductiveaxis. 54. 2302–2310. (doi:10.1186/1471-2202-5-54) et al

92 (doi:10.1073/pnas.91.1.395)

(doi:10.1159/000452105) 263–271. 5

. 2016Defininganovelleptin-melanocortin-kisspeptin (doi:10.1210/en.138.3.1190) 36 844–857.

134 Journal ofNeuroendocrinology Journal 2336–2346. Nature (doi:10.1210/en.2011-0096) 1718–1722. Endocrinology Endocrinology (doi:10.1016/j.physbeh.2007.05.021)

428 Scientific Reports (doi:10.1016/j.molmet.2016.08.003) 569–574. (doi:10.1111/j.1460-9568.2012.08168.x) Biology ofReproduction

297 (doi:10.1210/en.134.4.1718) American Journal ofPhysiology: American Journal

et al E1247–E1259. 141 151 (doi:10.1038/nature02440) . 2004AMP-kinaseregulates 752–762. 1863–1871. m

5

c 16176. © 2017SocietyforEndocrinology

Endocrinology evans

Neuroendocrinology 24 1412–1422. and (doi:10.1210/ (doi:10.1152/ European Journal of European Journal (doi:10.1038/ Physiology and Printed inGreatBritain

(doi:10.1210/ 46 BMC Neuroscience g 1151–1157.

Molecular 138 m Endocrinology

PNAS anderson

.

91

Quennell JH,MulliganAC,Tups A,LiuX,PhippsSJ,KempCJ, Qiu X,DaoH,Wang M,HestonA,Garcia KM,SangalA,DowlingAR, Qi Y, InoueK, FuM,InuiA&HerzogH2015Chronicoverproduction Poling MC,ShiehMP, Munaganuru N,LuoE&KauffmanAS2014 Pinilla L,Fernandez-FernandezR,RoaJ,CastellanoJM,Tena-Sempere M Pinilla L,Fernandez-FernandezR,Vigo E,NavarroVM,RoaJ, Ollmann MM,Wilson BD,Yang YK,KernsJA,ChenY, GantzI& Ojeda SR,LomnicziA&SandauUS2008Glial-gonadotrophinhormone O’Byrne KT, LunnSF&DixsonAF1988Effectsofacutestressonthe Niswender KD,GallisB,BlevinsJE,CorsonMA,SchwartzMW&Baskin DG Niswender KD,MortonGJ,StearnsWH,RhodesCJ,MyersMGJr& Niswender KD&SchwartzMW2003Insulinandleptinrevisited: Nikolarakis KE,LoefflerJP, AlmeidaOF&HerzA1988Pre-and Ping L,MaheshVB,BhatGK&BrannDW1997Regulationof Ovesjo ML,GamstedtM,CollinM&MeisterB2001GABAergicnature Olson VG&NestlerEJ2007Topographical organizationofGABAergic of reproduction Nutritional regulation Published byBioscientifica Ltd. Endocrinology regulates gonadotropin-releasinghormone neuronalfunction. GrattanDR&Anderson GM2009Leptinindirectly Herbison AE, period. signaling interactinthemousekiss1neuronduringperipubertal Faulkner LD,MolitorSC,EliasCF, npep.2015.02.002) adulthood. challenge onRFRP-3neuronsofmiceindevelopmentand Examination oftheinfluenceleptinandacutemetabolic (doi:10.1152/ajpendo.00274.2007) ofPhysiology:EndocrinologyandMetabolism Journal Y2 inthecontrolofgonadotropinsecretionrat. & AguilarE2007SelectiveroleofneuropeptideYreceptorsubtype Barsh GS 1997Antagonismofcentralmelanocortinreceptorsin Barsh GS (doi:10.1111/j.1365-2826.2008.01712.x) control ofGnRHsecretion. (GnRH) neuroneinteractionsinthemedianeminenceand joe.0.1180259) jacchus). patterns ofLHsecretioninthecommonmarmoset(Callithrix Cytochemistry activation byinsulinandleptin. 2003 Immunocytochemicaldetectionofphosphatidylinositol3-kinase induced anorexia. Schwartz MW2001Intracellularsignalling.Keyenzymeinleptin- (doi:10.1016/S0091-3022(02)00105-X) signaling capabilities. adiposity signalswithoverlappingphysiologicalandintracellular 677–683. intake andbodyweight. of ghrelininthehypothalamusleadstotemporalincreasefood and Metabolism potentiated infastedrats. effectofPYY-(3-36)Stimulatory ongonadotropinsecretionis PinedaR,Tena-SempereCastellano JM, M&AguilarE2006 Neuroendocrinology receptors inthesteroid-inducedluteinizinghormonesurge. by AMPA receptors.EvidenceforaphysiologicalroleofAMPA gonadotropin-releasing hormoneandluteinizingsecretion (doi:10.1046/j.1365-2826.2001.00662.x) nucleus. of hypothalamicleptintargetneuronesintheventromedialarcuate 87–95. neurons withintheventraltegmentalareaofrat. (doi:10.1126/science.278.5335.135) vitro andinvivobyagouti-relatedprotein. gonadotropin-releasing hormone(GnRH). postsynaptic actionsofGABAonthereleasehypothalamic (doi:10.1002/syn.20345) PLoS ONE Journal ofNeuroendocrinology Journal Journal ofEndocrinology Journal (doi:10.1016/0361-9230(88)90208-0) Neuroendocrinology

51 150

290 275–283.

2805–2812.

10 Nature 66 E1162–E1171. e0121974. 246–253. Frontiers inNeuroendocrinology

Neuropeptides 413 (doi:10.1177/002215540305100302) American Journal of Physiology: Endocrinology ofPhysiology:Endocrinology American Journal Journal ofNeuroendocrinology Journal Downloaded fromBioscientifica.com at09/24/202102:55:05PM

794–795. 100 (doi:10.1210/en.2008-1693) Journal of Histochemistry and and ofHistochemistry Journal (doi:10.1159/000127245)

(doi:10.1371/journal.pone.0121974) 118 317–333. et al (doi:10.1152/ajpendo.00469.2005)

259–264. 13

50 . 2015Insulinandleptin (doi:10.1038/35101657) 505–516. 23–28. Brain Research Bulletin Science (doi:10.1159/000369276) 58 (doi:10.1677/ : 2 (doi:10.1016/j.

293

278

24 Synapse E1385–E1392. 1–10. 135–138. American

20 732–742. R126

61

21 via freeaccess

Review m c evans and g m anderson Nutritional regulation 58:2 R127 of reproduction

Quennell JH, Howell CS, Roa J, Augustine RA, Grattan DR & Schwartz MW, Marks JL, Sipols AJ, Baskin DG, Woods SC, Kahn SE & Anderson GM 2011 Leptin deficiency and diet-induced obesity Porte D Jr 1991 Central insulin administration reduces neuropeptide reduce hypothalamic kisspeptin expression in mice. Endocrinology Y mRNA expression in the arcuate nucleus of food-deprived lean (Fa/ 152 1541–1550. (doi:10.1210/en.2010-1100) Fa) but not obese (fa/fa) Zucker rats. Endocrinology 128 2645–2647. Ramamurthy S & Ronnett GV 2006 Developing a head for energy (doi:10.1210/endo-128-5-2645) sensing: AMP-activated as a multifunctional metabolic Sheffer-Babila S, Sun Y, Israel DD, Liu SM, Neal-Perry G & Chua SC Jr sensor in the brain. Journal of Physiology 574 85–93. (doi:10.1113/ 2013 Agouti-related peptide plays a critical role in leptin’s effects on jphysiol.2006.110122) female puberty and reproduction. American Journal of Physiology: Rizwan MZ, Harbid AA, Inglis MA, Quennell JH & Anderson GM 2014 Endocrinology and Metabolism 305 E1512–E1520. (doi:10.1152/ Evidence that hypothalamic RFamide related peptide-3 neurones are ajpendo.00241.2013) not leptin-responsive in mice and rats. Journal of Neuroendocrinology Shi H, Sorrell JE, Clegg DJ, Woods SC & Seeley RJ 2010 The roles of 26 247–257. (doi:10.1111/jne.12140) leptin receptors on POMC neurons in the regulation of sex-specific Roa J & Herbison AE 2012 Direct regulation of GnRH neuron energy homeostasis. Physiology & Behavior 100 165–172. excitability by arcuate nucleus POMC and NPY neuron (doi:10.1016/j.physbeh.2010.02.018) neuropeptides in female mice. Endocrinology 153 5587–5599. Shioda S, Kageyama H, Takenoya F & Shiba K 2011 Galanin-like peptide: (doi:10.1210/en.2012-1470) a key player in the homeostatic regulation of feeding and energy Roa J & Tena-Sempere M 2014 Connecting metabolism and metabolism? International Journal of Obesity 35 619–628. reproduction: roles of central energy sensors and key molecular (doi:10.1038/ijo.2010.202) mediators. Molecular and Cellular Endocrinology 397 4–14. Singireddy AV, Inglis MA, Zuure WA, Kim JS & Anderson GM 2013 (doi:10.1016/j.mce.2014.09.027) Neither signal transducer and activator of transcription 3 (STAT3) or Roa J, Garcia-Galiano D, Varela L, Sanchez-Garrido MA, Pineda R, STAT5 signaling pathways are required for leptin’s effects on fertility Castellano JM, Ruiz-Pino F, Romero M, Aguilar E, Lopez M, et al. in mice. Endocrinology 154 2434–2445. (doi:10.1210/en.2013-1109) 2009 The mammalian target of rapamycin as novel central regulator Skrapits K, Kanti V, Savanyu Z, Maurnyi C, Szenci O, Horvath A, Borsay BA, of puberty onset via modulation of hypothalamic Kiss1 system. Herczeg L, Liposits Z & Hrabovszky E 2015 Lateral hypothalamic orexin Endocrinology 150 5016–5026. (doi:10.1210/en.2009-0096) and melanin-concentrating hormone neurons provide direct input to Rodriguez EM, Blazquez JL, Pastor FE, Pelaez B, Pena P, Peruzzo B & gonadotropin-releasing hormone neurons in the human. Frontiers in Amat P 2005 Hypothalamic tanycytes: a key component of brain- Cellular Neuroscience 9 348. (doi:10.3389/fncel.2015.00348) endocrine interaction. International Review of Cytology 247 89–164. Smith JT 2009 Sex steroid control of hypothalamic Kiss1 expression in (doi:10.1016/S0074-7696(05)47003-5) sheep and rodents: comparative aspects. Peptides 30 94–102. Roland AV & Moenter SM 2011 Regulation of gonadotropin-releasing (doi:10.1016/j.peptides.2008.08.013) hormone neurons by glucose. Trends in Endocrinology and Metabolism Smith JT, Acohido BV, Clifton DK & Steiner RA 2006 KiSS-1 neurones 22 443–449. (doi:10.1016/j.tem.2011.07.001) are direct targets for leptin in the ob/ob mouse. Journal of Sandau US, Mungenast AE, Alderman Z, Sardi SP, Fogel AI, Taylor B, Neuroendocrinology 18 298–303. Parent AS, Biederer T, Corfas G & Ojeda SR 2011a SynCAM1, a (doi:10.1111/j.1365-2826.2006.01417.x) synaptic adhesion molecule, is expressed in astrocytes and contributes Souter I, Baltagi LM, Kuleta D, Meeker JD & Petrozza JC 2011 Women, to erbB4 receptor-mediated control of female sexual development. weight, and fertility: the effect of body mass index on the outcome Endocrinology 152 2364–2376. (doi:10.1210/en.2010-1435) of superovulation/intrauterine insemination cycles. Fertility and Sandau US, Mungenast AE, McCarthy J, Biederer T, Corfas G & Ojeda SR Sterility 95 1042–1047. (doi:10.1016/j.fertnstert.2010.11.062) 2011b The synaptic cell adhesion molecule, SynCAM1, mediates Spanswick D, Smith MA, Mirshamsi S, Routh VH & Ashford ML 2000 Journal of Molecular Endocrinology astrocyte-to-astrocyte and astrocyte-to-GnRH neuron adhesiveness in Insulin activates ATP-sensitive K+ channels in hypothalamic neurons the mouse hypothalamus. Endocrinology 152 2353–2363. of lean, but not obese rats. Nature Neuroscience 3 757–758. (doi:10.1210/en.2010-1434) (doi:10.1038/77660) Sanz E, Quintana A, Deem JD, Steiner RA, Palmiter RD & McKnight GS Stoyanovitch AG, Johnson MA, Clifton DK, Steiner RA & Fraley GS 2005 2015 Fertility-regulating Kiss1 neurons arise from hypothalamic Galanin-like peptide rescues reproductive function in the diabetic POMC-expressing progenitors. Journal of Neuroscience 35 5549–5556. rat. Diabetes 54 2471–2476. (doi:10.2337/diabetes.54.8.2471) (doi:10.1523/JNEUROSCI.3614-14.2015) Sullivan SD & Moenter SM 2004 Gamma-aminobutyric acid neurons Sato I, Arima H, Ozaki N, Watanabe M, Goto M, Hayashi M, Banno R, integrate and rapidly transmit permissive and inhibitory metabolic Nagasaki H & Oiso Y 2005 Insulin inhibits neuropeptide Y gene cues to gonadotropin-releasing hormone neurons. Endocrinology 145 expression in the arcuate nucleus through GABAergic systems. 1194–1202. (doi:10.1210/en.2003-1374) Journal of Neuroscience 25 8657–8664. (doi:10.1523/ Sullivan SD, DeFazio RA & Moenter SM 2003 Metabolic regulation of JNEUROSCI.2739-05.2005) fertility through presynaptic and postsynaptic signaling to Schaeffer M, Langlet F, Lafont C, Molino F, Hodson DJ, Roux T, gonadotropin-releasing hormone neurons. Journal of Neuroscience 23 Lamarque L, Verdie P, Bourrier E, Dehouck B, et al. 2013 Rapid 8578–8585. sensing of circulating ghrelin by hypothalamic appetite-modifying Sun YX, Ahmed S & Smith RG 2003 Deletion of ghrelin impairs neither neurons. PNAS 110 1512–1517. (doi:10.1073/pnas.1212137110) growth nor appetite. Molecular and Cellular Biology 23 7973–7981. Scheid JL & De Souza MJ 2010 Menstrual irregularities and energy (doi:10.1128/MCB.23.22.7973-7981.2003) deficiency in physically active women: the role of ghrelin, PYY and Sun YX, Butte NF, Garcia JM & Smith RG 2008 Characterization of adult adipocytokines. Medicine and Sport Science 55 82–102. ghrelin and ghrelin receptor knockout mice under positive and (doi:10.1159/000321974) negative energy balance. Endocrinology 149 843–850. (doi:10.1210/ Schneider LF & Warren MP 2006 Functional hypothalamic amenorrhea en.2007-0271) is associated with elevated ghrelin and disordered eating. Fertility and Takatsu Y, Matsumoto H, Ohtaki T, Kumano S, Kitada C, Onda H, Sterility 86 1744–1749. (doi:10.1016/j.fertnstert.2006.05.051) Nishimura O & Fujino M 2001 Distribution of galanin-like peptide Schneider LF, Monaco SE & Warren MP 2008 Elevated ghrelin level in in the rat brain. Endocrinology 142 1626–1634. (doi:10.1210/ women of normal weight with amenorrhea is related to disordered endo.142.4.8089) eating. Fertility and Sterility 90 121–128. (doi:10.1016/j. Takenoya F, Guan JL, Kato M, Sakuma Y, Kintaka Y, Kitamura Y, fertnstert.2007.06.002) Kitamura S, Okuda H, Takeuchi M, Kageyama H, et al. 2006 Neural

http://jme.endocrinology-journals.org © 2017 Society for Endocrinology Published by Bioscientifica Ltd. DOI: 10.1530/JME-16-0212 Printed in Great Britain Downloaded from Bioscientifica.com at 09/24/2021 02:55:05PM via free access Journal of Molecular Endocrinology Wattez JS,DelahayeF, LukaszewskiMA,RisoldPY, EberleD,Vieau D& Wang J&LeibowitzKL1997Centralinsulininhibitshypothalamic Wan Q,XiongZG,ManHY, AckerleyCA,BrauntonJ,LuWY, BeckerLE, Wade GN&JonesJE2004Neuroendocrinologyofnutritional infertility. Vulliemoz NR,XiaoE,Xia-ZhangL,Wardlaw SL&FerinM2005Central Volkow ND,Wang GJ&BalerRD2011Reward,dopamineandthe van deWall E,LeshanR,XuAW, BalthasarN,CoppariR,LiuSM,JoYH, True C,Verma S,GroveKL&SmithMS2013Cocaine-and Trinko R,GanGL,GaoXB,SearsRM,GuarnieriDJ&DiLeoneRJ2011 Tong QC,Ye CP, McCrimmonRJ,DhillonH,ChoiB,KramerMD,Yu J, Tong Q,Ye C,McCrimmonRJ,DhillonH,ChoiB,KramerMD,Yu J, Tilbrook AJ,Turner AI&ClarkeIJ2002Stressandreproduction: central Tilbrook AJ,Turner AI&ClarkeIJ2000Effectsofstressonreproduction Tena-Sempere M2006KiSS-1andreproduction:focusonitsroleinthe DOI: 10.1530/JME-16-0212 http://jme.endocrinology-journals.org Review Breton C2013Perinatalnutritionprogramsthehypothalamic 8993(97)00963-3) intact rats. galanin andneuropeptideYgeneexpressionpeptidereleasein (doi:10.1038/41792) receptors topostsynapticdomainsbyinsulin. MacDonald JF&Wang YT1997Recruitmentoffunctional GABA(A) Physiology ofPhysiology:Regulatory,American Journal IntegrativeandComparative 146 hormone releaseintheovariectomizedrhesusmonkey. infusion ofagouti-relatedpeptidesuppressespulsatileluteinizing Sciences control offoodintake:implicationsforobesity. controlling metabolismandingestion. individual functionsofleptinreceptormodulatedneurons MacKenzie RG,AllisonDB,DunNJ, 2821–2832. induced reproductiveinhibitioninfemales. and kisspeptincellsmaycontributetonegativeenergybalance- amphetamine-regulated transcriptisapotentstimulatorofGnRH area. Erk1/2 mediatesleptinreceptorsignalingintheventraltegmental 383–393. neurocircuitry thatpreventshypoglycemia. release byventromedialhypothalamicneuronsispartofthe Yang ZF, ChristiansenLM,LeeCE, 383–393. neurocircuitry thatpreventshypoglycemia. release byventromedialhypothalamicneuronsispartofthe Yang Z,ChristiansenLM,LeeCE, 83–100. mechanisms andsexdifferencesinnon-rodentspecies. ror.0.0050105) differences. in non-rodentmammals:theroleofglucocorticoidsandsex (doi:10.1159/000095549) metabolic regulationoffertility. 2885–2893. hormone-releasing hormone(LHRH)-containingneurons. interaction betweengalanin-likepeptide(GALP)-andluteinizing 784–789. PLoS ONE

15 (doi:10.1080/10253890290027912)

(doi:10.1016/j.cmet.2007.04.001) (doi:10.1016/j.cmet.2007.04.001) 287 37–46. Brain Research Reviews ofReproduction (doi:10.1210/en.2013-1156) (doi:10.1016/j.peptides.2006.05.012) R1277–R1296. (doi:10.1210/en.2004-1093)

6 e27180. (doi:10.1016/j.tics.2010.11.001)

777 (doi:10.1371/journal.pone.0027180) 231–236. (doi:10.1152/ajpregu.00475.2004) Neuroendocrinology et al

et al 5 105–113. et al m . 2007 . 2007 Endocrinology

(doi:10.1016/S0006- c . 2008Collectiveand © 2017SocietyforEndocrinology

evans Cell Metabolism Cell Metabolism Endocrinology a b Synapticglutamate Nature Synapticglutamate (doi:10.1530/ and Trends inCognitive Printed inGreatBritain

83

149 388 g 275–281.

m Stress Endocrinology 1773–1785.

686–690. 154 anderson Peptides

5 5

5

27

Wu Q,WhiddonBB&PalmiterRD2012Ablationofneuronsexpressing Wu M,DumalskaI,MorozovaE,vandenPolA&AlrejaM2009 Wittmann G,HrabovszkyE&LechanRM2013Distinctglutamatergic Welt CK,ChanJL,BullenJ,MurphyR,SmithP, DePaoliAM,KaralisA& Accepted Preprint published online 5 January 2017 Accepted Preprintpublishedonline5January 2017 Accepted 5January Received infinalform23December2016 Yang L,QiY&Yang Y2015Astrocytescontrolfoodintakebyinhibiting Williams KW, MargathoLO,LeeCE,ChoiM,S,ScottMM,EliasCF Williams NI,BergaSL&CameronJL2007Synergismbetween Zuure WA, Quennell JH&AndersonGM2016Leptinresponsiveand Zuure WA, Roberts AL,QuennellJH&AndersonGM2013Leptin Zhu CC,ZhangH,JS,LiZ,ZhaoJ,W&YQ2013 Zhang C,BoschMA,LevineJE,RonnekleivOK&KellyMJ2007 Zhang Y, ProencaR,MaffeiM,BaroneLeopoldL&FriedmanJM Zhang X&vandenPolAN2016Hypothalamicarcuate nucleustyrosine Yi CX,HabeggerKM,ChowenJA,SternJ&Tschop MH2011Arolefor of reproduction Nutritional regulation Published byBioscientifica Ltd. agouti-related protein,butnotmelaninconcentratinghormone,in PNAS blocks kisspeptinactivation,linkingenergybalancetoreproduction. Melanin-concentrating hormonedirectlyinhibitsGnRHneuronsand ofComparativeNeurology Journal neurons revealedbyinsituhybridizationmaleratsandmice. and GABAergicsubsetsofhypothalamicpro-opiomelanocortin JNEUROSCI.3118-09.2010) of Neuroscience distinct populationsofarcuate proopiomelanocortinneurons. & ElmquistJK2010Segregationofacuteleptinandinsulineffectsin and Metabolism in cynomolgusmonkeys. psychosocial andmetabolicstressors:impactonreproductivefunction 987–997. hypothalamic amenorrhea. Mantzoros CS2004Recombinanthumanleptininwomenwith 980–990. melanocortin systeminoffspring. GABAergic projectionstotherostralpreopticareainmice. (doi:10.1523/JNEUROSCI.2278-13.2013) for reproductivefunction. signaling inGABAneurons,butnotglutamateisrequired (doi:10.1016/j.fertnstert.2012.11.022) of maleob/obmouse. Inhibition ofghrelinsignalingimprovesthereproductivephenotype (doi:10.1523/JNEUROSCI.1657-07.2007) metabolic inhibition. that areregulatedbyestrogenandresponsivetoglucose Gonadotropin-releasing hormoneneuronsexpressK(ATP) channels homologue. 1994 Positionalcloningofthemouseobesegeneanditshuman Nature Neuroscience hydroxylase neuronsplayorexigenicroleinenergyhomeostasis. 93 astrocytes inthecentralcontrolofmetabolism. 798–807. AGRP neuronactivityviaadenosineA1receptors. 109 leptin-deficient micerestoresmetabolicfunctionsandfertility. Neuroendocrinology 143–149. 3155–3160.

106 (doi:10.1056/NEJMoa040388) (doi:10.1055/s-0033-1357182) (doi:10.1016/j.celrep.2015.04.002) 17217–17222. Nature (doi:10.1159/000324888)

30 293 (doi:10.1073/pnas.1120501109) 2472–2479.

28

E270–E276. 19 372 12357. Journal ofNeuroscience Journal 1341–1347. Fertility andSterility Fertility

425–432. (doi:10.1073/pnas.0908200106) American Journal of Physiology: Endocrinology ofPhysiology:Endocrinology American Journal Journal ofNeuroscience Journal Downloaded fromBioscientifica.com at09/24/202102:55:05PM New England Journal ofMedicine New EnglandJournal (doi:10.1111/jne.12357) (doi:10.1523/

(doi:10.1152/ajpendo.00108.2007) 521 Hormone andMetabolicResearchHormone (doi:10.1038/nn.4372) 3287–3302.

99

27 58 918–926. 10153–10164. : 2 Neuroendocrinology

33 Cell Reports 17874–17883.

Journal of Journal 351 R128

11 Journal Journal PNAS

45 via freeaccess