<<

Molecular Psychiatry (1997) 2, 355–358  1997 Stockton Press All rights reserved 1359–4184/97 $12.00

REVIEW ARTICLE: A TRIBUTE TO SM McCANN The transforming alpha family is involved in the neuroendocrine control of mammalian puberty SR Ojeda, YJ Ma and F Rage

Division of , Oregon Regional Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA

The concept is proposed that the central control of mammalian female puberty requires the interactive participation of neuronal networks and glial cells of the astrocytic lineage. Accord- ing to this concept and control the pubertal process by regulating the secretory activity of those neurons that secrete luteinizing -releasing hormone (LHRH). LHRH, in turn, governs sexual development by stimulating the secretion of pituitary gonadotropins. Astrocytes affect LHRH neuronal function via a mechanism involving several growth factors and their corresponding receptors. Our laboratory has ident- ified two members of the /transforming growth factor (EGF/TGF␣) fam- ily as components of the glial-neuronal interactive process that regulates LHRH secretion. Transforming growth factor alpha (TGF␣) and its distant congener neu-differentiation factor, NDF, are produced in hypothalamic astrocytes and stimulate LHRH release via a glial interme- diacy. The actions of TGF␣ and NDF on hypothalamic astrocytes involve the interactive acti- vation of their cognate receptors and the synergistic effect of both ligands in stimulating the

glial release of E2 (PGE2). In turn, PGE2 acts directly on LHRH neurons to stimu- late LHRH release. A variety of experimental approaches has led to the conclusion that both TGF␣ and NDF are physiological components of the central mechanism controlling the initiation of female puberty. Keywords: glial cells; ; growth factors; sexual development; astrocytes; cell–cell com- munication; LHRH neurons

The onset of mammalian female puberty requires the ronal network is well-documented, recent studies have functional activation of a specialized group of hypo- raised the possibility that glial cells may also play a thalamic neurons that produce - role in the control of LHRH neuronal activity.9 Ana- releasing hormone (LHRH), the neurohormone govern- tomical evidence demonstrates that the glial ensheath- ing sexual maturation and reproductive function. ment of LHRH neurons in both primates and rodents Elucidation of the mechanisms controlling LHRH neu- is substantial,10,11 the main difference between species ronal function is critical for the understanding of the being a more prominent glial apposition to LHRH cellular and molecular processes underlying sexual perikarya in primates.10 Glial apposition of LHRH development and adult reproductive capacity. It is well nerve terminals in the median eminence of the hypo- accepted that transsynaptic inputs, both excitatory1,2 thalamus is, however, similarly abundant in both rats and inhibitory,3 contribute to the central regulation of and primates. The median eminence contains astro- LHRH secretion. A combination of a decrease in trans- cytes and modified ependymoglial cells known as tany- synaptic inhibitory tone and an increase in excitatory cytes.12 These specialized glial cells line the ventral inputs to LHRH neurons is thought to be instrumental portion of the third ventricle and send their processes in bringing about the initiation of puberty. The main towards the endothelial wall of the portal blood vascul- excitatory inputs affecting LHRH secretion include ature, establishing along their way intimate contact neuronal systems that utilize (NE),4 with LHRH nerve terminals.12,13 More intriguingly, sex Y (NPY)1 and excitatory amino acids.2,5,6 steroids appear to modulate this structural arrange- The inhibitory , on the other hand, ment, as gonadectomy decreases the space between are gamma aminobutyric acid (GABA)3,7 and opioid LHRH nerve terminals and the basal lamina, presum- .8 ably due to retraction of the glial cells.14 An obvious While this transsynaptic regulation of the LHRH neu- interpretation of these structural changes is that they increase the access of LHRH nerve endings to the portal vessels. The finding that about 50% of glial cells in 15 Correspondence: SR Ojeda, DVM, Division of Neuroscience, Ore- the median eminence have receptors further gon Regional Primate Research Center, 505 NW 185th Avenue, supports the notion of a direct action of estrogen on Beaverton, OR 97006, USA. E-mail: ojedasȰohsu.edu hypothalamic astrocytes.16 As glial-neuronal synaptoid Glial growth factors and LHRH secretion SR Ojeda et al 356 specializations are commonly seen in the median emi- logical blockade of EGFR activity36 tar- nence,12 this region appears to be a primary site where geted to the median eminence delays the onset of glial cells influence LHRH neuronal function. In this puberty,23 suggesting that activation of EGFR in this article, we will discuss some of the recent results region of the hypothalamus is a physiological compo- obtained by our laboratory concerning the potentially nent of the central mechanism underlying the acqui- important role played by glial cells in the regulation of sition of reproductive competence. In another study, LHRH secretion. we asked the question as to whether focal activation In a series of reports, we documented the concept of TGF␣ secretion would be able to stimulate LHRH that glia-derived transforming growth factor alpha neuronal activity, and thus accelerate the onset of (TGF␣), a mitogenic polypeptide member of the epider- puberty. To produce this focal increase we used two mal growth factor (EGF) family,17–19 plays an important gene delivery systems. In one of them, fibroblasts were role in the process by which astrocytes affect LHRH stably transfected with a retroviral construct in which secretory activity.16,20–24 More recently, we obtained expression of the TGF␣ gene was directed by evidence for the involvement of other members of the the phosphoglycerate kinase promoter, which is consti- EGF/TGF␣ family in this process. , also tutively active in most mammalian cells. The other known as heregulins25 or—in the case of the rat—as delivery system consisted of a plasmid construct in neu-differentiation factor (NDF),26 are alternatively which the human TGF␣ gene was placed under the spliced products of a single gene.26,27 Our studies control of a mouse MT promoter. Since this promoter showed that NDF is produced in hypothalamic astro- is activated by heavy metals, TGF␣ expression can be cytes and facilitates LHRH release through a mech- increased several-fold by simply exposing the cells car-

anism involving stimulation of glial PGE2 release and rying the construct to zinc chloride. facilitation of TGF␣ action on astrocytes (Ma et al, The genetically modified fibroblasts were then unpublished). implanted into different hypothalamic sites of juvenile The biological actions of TGF␣ and NDF are initiated female rats and the onset of puberty was evaluated. To by binding to membrane-spanning receptors endowed activate a circadian expression of the TGF␣ gene con- with kinase activity for cellular signaling.28–32 trolled by the MT promoter, animals bearing these cells The for TGF␣ (which also recognizes EGF, were injected once daily with zinc chloride. The amphiregulins and other related members of the results demonstrated that cells carrying either con- family) is a 170-kDa glycosylated protein known as the struct were able to accelerate the onset of puberty only EGF receptor (EGFR). EGFR has an extracellular when they were grafted in the immediate vicinity of domain containing the motifs essential for LHRH neurons. The effect appears to be more robust recognition, a single transmembrane domain, when the cells were located in contact with LHRH and an intracellular domain endowed with tyrosine nerve terminals in the median eminence of the hypo- kinase activity. The receptors for NDF, known as HER- thalamus.37 Thus, these experiments demonstrate that 3 (also named ERBB-3)29,31 and HER-4,30 are related to a localized activation of TGF␣ production can acceler- EGFR. An additional related receptor, known as HER- ate puberty, as long as the activation occurs within the 2 (neu) receptor,32,33 appears to function as an auxiliary confines of the LHRH microenvironment. subunit for the high affinity binding of TGF␣/EGF to As indicated before, TGF␣ is not the only member of EGFR and NDF/neuregulins to HER-3 and HER-4.34 the EGF family that participates in the control of LHRH In the hypothalamus of both rats23 and nonhuman secretion. In very recent in vitro experiments we found primates,20 the TGF␣ gene and its protein product are that glia-derived NDF enhances the stimulatory action predominantly expressed in astroglial cells of certain of TGF␣ on LHRH release via activation of an HER- nuclei, such as the suprachiasmatic nucleus, arcuate 2/HER-4 receptor complex. Blockade of NDF signal nucleus and dorsomedial nucleus.23 Both TGF␣ mRNA transduction via an antisense oligonucleotide, which and protein are also abundant in tanycytes and glial disrupts the synthesis of HER-2, also attenuated the cells of the median eminence, the level of expression effect of TGF␣ on LHRH release indicating that the increasing significantly around the time of puberty.20,23 TGF␣/EGFR and NDF/HER ligand-receptor systems Importantly, EGFR is also present in the median emi- function in a tightly coordinated manner to regulate nence,20,22 indicating the involvement of a TGF␣-EGFR neuroendocrine reproductive function. interaction in neuroendocrine regulation. That TGF␣ The stimulatory effects of TGF␣ on LHRH neuronal may be involved in the control of LHRH neuronal activity are not exerted directly, as LHRH neurons do activity was demonstrated by the finding that TGF␣ is not contain EGFR.22 Instead, TGF␣ acts on hypothal- able to stimulate the release of LHRH from median emi- amic astrocytes, which are endowed with EGFR,16,38 nence fragments in a dose-related manner.35 Several and respond to the with an increase in TGF␣ ␣ 16 39 observations suggest that TGF -induced activation of and PGE2 release. The conditioned EGFR contributes to the normal initiation of mam- medium from hypothalamic astrocytes treated with malian female puberty. For instance, overexpression of TGF␣ stimulates LHRH release from an LHRH-produc- TGF␣ in transgenic mice carrying a human TGF␣ gene ing cell line, which expresses no EGFR and is unre- ␣ 38 under the control of a metallothionein (MT) promoter sponsive to TGF itself. That glial PGE2 is, at least in (which is activated by heavy metals, such as Zn2+) part, responsible for this stimulatory effect is suggested 21 induces precocious puberty. In contrast, pharmaco- by the finding that blockade of PGE2 action by immu- Glial growth factors and LHRH secretion SR Ojeda et al 357

Figure 1 Glial–neuronal interactive mechanisms proposed to participate in the central regulation of mammalian puberty. While neuronal networks associated with LHRH neurons act transsynaptically [1] to regulate LHRH secretion, they can also affect astrocytic function via specific receptors located on glial cells [2]. Excitatory amino acids exemplify this regulatory influence as they can affect glial production of growth factors via metabotropic receptors. Astrocytes affect LHRH secretion through both direct actions on LHRH neurons (for instance, fibroblast growth factor),38 and indirect paracrine/autocrine mech- anisms [3] leading to the formation and release of neuroactive molecules able to directly affect LHRH release, such as prostaglan- 41 din E2 [4]. PGE2 is also able to affect astrocytic function. Growth factors and other neuroactive substances produced by astrocytes may also influence the transsynaptic communication between LHRH neurons and its associate neuronal circuitry [5]. noneutralization of the prostaglandin in the con- Acknowledgements ditioned medium abolishes the increase in LHRH induced by this medium.39 This work was supported by NIH grants HD25123, P30 In addition to TGF␣ itself, glial expression of the Population Center Grant HD18185, and RR00163 for TGF␣ gene appears to be upregulated by gonadal ster- the operation of the ORPRC. oids (estrogen) and neuronal inputs (excitatory amino 16,40 acids), suggesting that in addition to the above out- References lined glial–neuronal signaling pathway mediated by growth factors and , there is a neuron-to- 1 Crowley WR, Parker SL, Sahu A, Kalra SP. Interacting transmem- glia signaling mechanism mediated by excitatory neu- brane signals regulating GnRH and LH secretion. In: Plant TM, Lee PA (eds). The Neurobiology of Puberty. Journal of Endocrinology rotransmitters, and a hormone regulatory system Ltd: Bristol, UK, 1995, pp 41–54. effected by . Our experiments indicate that 2 Brann DW, Mahesh VB. Excitatory amino acids: function and sig- glutamate induces glial synthesis of TGF␣ via acti- nificance in reproduction and neuroendocrine regulation. Front vation of metabotropic receptors,40 thus highlighting Neuroendocrinol 1994; 15: 3–49. the importance of excitatory amino acids for both the 3 Terasawa E. Mechanisms controlling the onset of puberty in pri- mates: the role of GABAergic neurons. In: Plant TM, Lee PA (eds). transsynaptic and glial control of LHRH neuronal func- The Neurobiology of Puberty. Journal of Endocrinology Ltd: Bristol, tion during female sexual development. UK, 1995, pp 139–151. In summary (Figure 1), the initiation of mammalian 4 Ramirez VD, Feder HH, Sawyer CH. The role of brain catecholam- puberty requires a multiplicity of coordinated events ines in the regulation of LH secretion: a critical inquiry. In: Martini L, Ganong WF (eds). Frontiers in Neuroendocrinology, Vol 8. Raven that ultimately enhance the secretory activity of the Press: New York, 1984, pp 27–84. LHRH neuronal network. These events appear to 5 Ojeda SR. The mystery of mammalian puberty: how much more include: (a) an increase in transsynaptic excitatory do we know? Perspect Biol Med 1991; 34: 365–383. inputs to LHRH neurons mediated by neurotransmit- 6 Plant TM. Puberty in primates. In: Knobil E, Neill J (eds). The ters such as excitatory amino acids, NE and NPY; (b) a Physiology of Reproduction, 2nd edn, Vol 2. Raven Press: New York, 1994, pp 453–485. decrease in the transsynaptic GABA inhibitory regu- 7 Masotto C, Negro-Vilar A. Activation of gamma-amino butyric acid lation; and (c) an enhanced glial production of growth B-receptors abolishes naloxone-stimulated luteinizing hormone factors of the EGF family. These peptides appear to release. Endocrinology 1987; 121: 2251–2255. facilitate LHRH secretion via paracrine/autocrine inter- 8 Bhanot R, Wilkinson M. Opiatergic control of gonadotropin secretion during puberty in the rat. Endocrinology 1983; 113: actions involving the coordinated activation of related 596–602. tyrosine kinase receptors and the release of glial bioac- 9 Ojeda SR, Ma YJ. Neuroendocrine control of female puberty: the tive molecules able to directly stimulate LHRH release. role of brain growth factors. In: Sarkar DK, Barnes CD (eds). Repro- Glial growth factors and LHRH secretion SR Ojeda et al 358 ductive Neuroendocrinology of Aging and Drug Abuse. CRC Press: O, Kirk C, Hendricks M, Danehy F, Misumi D, Sudhalter J, Kobaya- Boca Raton, FL, 1995, pp 41–58. shi K, Wroblewski D, Lynch C, Baldassare M, Hiles I, Davis JB, 10 Witkin JW, Ferin M, Popilskis SJ, Silverman A. Effects of gonadal Hsuan JJ, Totty NF, Otsu M, McBurney RN, Waterfield MD, Stroob- steroids on the ultrastructure of GnRH neurons in the rhesus mon- ant P, Gwynne D. Glial growth factors are alternatively spliced key: synaptic input and glial apposition. Endocrinology 1991; 129: erbB2 ligands expressed in the nervous system. Nature 1993; 362: 1083–1092. 312–318. 11 Chen W-P, Witkin JW, Silverman AJ. Sexual dimorphism in the 28 Carpenter G. Receptors for epidermal growth factor and other poly- synaptic input to gonadotropin releasing hormone neurons. Endo- peptide . Ann Rev Biochem 1987; 56: 881–914. crinology 1990; 126: 695–702. 29 Kraus MH, Issing W, Miki T, Popescu NC, Aaronson SA. Isolation 12 Kobayashi H, Matsui T. Fine structure of the median eminence and and characterization of ERBB3, a third member of the its functional significance. In: Ganong WF, Martini L (eds). Fron- ERBB/epidermal family: evidence for over- tiers in Neuroendocrinology. Oxford University Press: New York, expression in a subset of human mammary tumors. Proc Natl Acad 1969, pp 3–46. Sci USA 1989; 86: 9193–9197. 13 Kozlowski GP, Coates PW. Ependymoneuronal specializations 30 Plowman GD, Culouscou J, Whitney GS, Green JM, Carlton GW, between LHRH fibers and cells of the cerebroventricular system. Foy L, Neubauer MG, Shoyab M. Ligand-specific activation of Cell Tissue Res 1985; 242: 301–311. HER4/p180erbB4, a fourth member of the epidermal growth factor 14 King JC, Letourneau RL. Luteinizing hormone-releasing hormone receptor family. Proc Natl Acad Sci USA 1993; 90: 1746–1750. terminals in the median eminence of rats undergo dramatic 31 Plowman GD, Whitney GS, Neubauer MG, Green JM, McDonald changes after gonadectomy, as revealed by electron microscopic VL, Todaro GJ, Shoyab M. Molecular cloning and expression of an image analysis. Endocrinology 1994; 134: 1340–1351. additional epidermal growth factor receptor-related gene. Proc Natl 15 Langub MC Jr, Watson RE Jr. Estrogen receptor-immunoreactive Acad Sci USA 1990; 87: 4905–4909. glia, endothelia, and ependyma in guinea pig preoptic area and 32 Schechter AL, Hung M, Vaidyanathan L, Weinberg RA, Yang-Feng median eminence: electron microscopy. Endocrinology 1992; 130: RL, Francke U, Ullrich A, Coussens L. The neu gene: an erb-B- 364–372. homologous gene distinct from and unlinked to the gene encoding 16 Ma YJ, Berg-von der Emde K, Moholt-Siebert M, Hill DF, Ojeda SR. the EGF receptor. Science 1985; 229: 976–978. Region-specific regulation of transforming growth factor ␣ (TGF␣) 33 Coussens L, Yang-Feng TL, Liao Y, Chen E, Gray A, McGrath J, gene expression in astrocytes of the neuroendocrine brain. J Neuro- Seeburg PH, Libermann TA, Schlessinger J, Francke U, Levinson sci 1994; 14: 5644–5651. A, Ullrich A. Tyrosine kinase receptor with extensive homology 17 Carpenter G, Cohen S. Epidermal growth factor. J Biol Chem 1990; to EGF receptor shares chromosomal location with neu . 165: 7709–7712. Science 1985; 230: 1132–1139. 18 Derynck R. Transforming growth factor ␣. Cell 1988; 54: 593–595. 34 Karunagaran D, Tsahar E, Beerli RR, Chen X. Graus-Porta D, Ratz- 19 Massague J. Transforming growth factor-␣. A model for membrane- kin BJ, Seger R, Hynes NE, Yarden Y. ErbB-2 is a common auxiliary anchored growth factors. J Biol Chem 1990; 265: 21393–21396. subunit of NDF and EGF receptors: implications for . 20 Ma YJ, Costa ME, Ojeda SR. Developmental expression of the EMBO J 1996; 15: 254–264. ␣ 35 Ojeda SR, Urbanski HF, Costa ME, Hill DF, Moholt-Siebert M. encoding transforming growth factor alpha (TGF ) and its receptor ␣ in the hypothalamus of female rhesus macaques. Neuroendocrinol- Involvement of transforming growth factor in the release of luteinizing-hormone releasing hormone from the developing ogy 1994; 60: 346–359. female hypothalamus. Proc Natl Acad Sci USA 1990; 87: 9698– 21 Ma YJ, Dissen GA, Merlino G, Coquelin A, Ojeda SR. Over- 9702. expression of a human transforming growth factor alpha (TGF␣) 36 Yaish P, Gazit A, Gilon C, Levitzki A. Blocking of EGF-dependent transgene reveals a dual antagonistic role of TGF␣ in female sexual by EGF receptor kinase inhibitors. Science 1988; development. Endocrinology 1994; 135: 1392–1400. 242: 933–935. 22 Ma YJ, Hill DF, Junier M, Costa ME, Felder SE, Ojeda SR. 37 Rage F, Hill DF, Serra-Esteves M, Breakefield XO, Coffey RJ, Costa Expression of epidermal growth factor receptor changes in the ME, McCann SM, Ojeda SR. Targeting transforming growth factor hypothalamus during the onset of female puberty. Mol Cell Neuro- ␣ expression to discrete loci of the neuroendocrine brain induces sci 1994; 5: 246–262. female sexual precocity. Proc Natl Acad Sci USA 1997; 2735–2740. 23 Ma YJ, Junier M, Costa ME, Ojeda SR. Transforming growth factor 38 Voigt P, Ma YJ, Gonzalez D, Fahrenbach WH, Wetzel WC, Berg-von ␣ alpha (TGF ) gene expression in the hypothalamus is developmen- der Emde K, Hill DF, Taylor KG, Costa ME, Seidah NG, Ojeda SR. tally regulated and linked to sexual maturation. Neuron 1992; 9: Neural and glial-mediated effects of growth factors acting via tyro- 657–670. sine kinase receptors on LHRH neurons. Endocrinology 1996; 137: 24 Ojeda SR. The neurobiology of mammalian puberty: has the contri- 2593–2605. bution of glial cells been underestimated? J NIH Res 1994; 6: 51–56. 39 Ma YJ, Berg-von der Emde K, Rage F, Wetsel WC, Ojeda SR. Hypo- 25 Holmes WE, Sliwkowski MX, Akita RW, Henzel WJ, Lee J, Park thalamic astrocytes respond to transforming growth factor alpha JW, Yansura D, Abadi N, Raab H, Lewis GD, Shepard HM, Kuang (TGF␣) with secretion of neuroactive substances that stimulate the W, Wood WI, Goeddel DV, Vandlen RL. Identification of heregulin, release of luteinizing hormone-releasing hormone (LHRH). Endo- a specific activator of p185erbB2. Science 1992; 256: 1205–1210. crinology 1997; 138: 19–25. 26 Wen D, Peles E, Cupples R, Suggs SV, Bacus SS, Luo Y, Trail G, 40 Ma YJ, Rage F, Ojeda SR. Regulation of transforming growth factor Hu S, Silbiger SM, Ben Levy R, Koski RA, Lu HS, Yarden Y. Neu alpha (TGF␣) gene expression in hypothalamic astrocytes by meta- differentiation factor: a transmembrane glycoprotein containing an botropic glutamate receptors. Soc Neurosci Abstr 1994; 20: 1070. EGF domain and an immunoglobulin homology unit. Cell 1992; 69: 41 Sawada M, Suzumura A, Ohno K, Marunouchi T. Regulation of ␣ 559–572. proliferation by prostaglandin E2 and the subtype of 27 Marchionni MA, Goodearl ADJ, Chen MS, Bermingham-McDonogh C. Brain Res 1993; 613: 67–73.