论文网首页|会计论文|管理论文|计算机论文|医药学|经济学论文|法学论文|社会学论文|文学论文|教育论文|理学论文|工学论文|艺术论文|哲学论文|文化论文|外语论文|论文格式
中国论文网

用户注册

设为首页

您现在的位置: 中国论文网 >> 医药学论文 >> 内科论文 >> 正文 会员中心
 药学论文   医学论文   临床医学论文   护理论文   口腔医学论文   肿瘤论文   妇产科学论文   内科论文   外科论文
 儿科论文   医学期刊
Minireview: The Neuroendocrine Regulation of Puberty: Is the Time Ripe for a Systems Biology Approach

【关键词】  puberty

    division of neuroscience (s.r.o., a.l., c.m., c.r., a.-s.p., v.m., a.e.m.), oregon national primate research center/oregon health and science university, beaverton, oregon 97006
    hospital for children and adolescents (s.h.), university of leipzig, 04317 leipzig, germany

    abstract

    the initiation of mammalian puberty requires an increase in pulsatile release of gnrh from the hypothalamus. this increase is brought about by coordinated changes in transsynaptic and glial-neuronal communication. as the neuronal and glial excitatory inputs to the gnrh neuronal network increase, the transsynaptic inhibitory tone decreases, leading to the pubertal activation of gnrh secretion. the excitatory neuronal systems most prevalently involved in this process use glutamate and the peptide kisspeptin for neurotransmission/neuromodulation, whereas the most important inhibitory inputs are provided by -aminobutyric acid (gaba)ergic and opiatergic neurons. glial cells, on the other hand, facilitate gnrh secretion via growth factor-dependent cell-cell signaling. coordination of this regulatory neuronal-glial network may require a hierarchical arrangement. one level of coordination appears to be provided by a host of unrelated genes encoding proteins required for cell-cell communication. a second, but overlapping, level might be provided by a second tier of genes engaged in specific cell functions required for productive cell-cell interaction. a third and higher level of control involves the transcriptional regulation of these subordinate genes by a handful of upper echelon genes that, operating within the different neuronal and glial subsets required for the initiation of the pubertal process, sustain the functional integration of the network. the existence of functionally connected genes controlling the pubertal process is consistent with the concept that puberty is under genetic control and that the genetic underpinnings of both normal and deranged puberty are polygenic rather than specified by a single gene. the availability of improved high-throughput techniques and computational methods for global analysis of mrnas and proteins will allow us to not only initiate the systematic identification of the different components of this neuroendocrine network but also to define their functional interactions.

    introduction

    in mammals, including humans, developmental changes in gonadotropin secretion are controlled by changes in pulsatile release of gnrh. at puberty, pulsatile gonadotropin secretion increases in a diurnal fashion, initially characterized in humans (1), but that also occurs in other species, including the rat (2). this change, necessary for normal gonadal development and function, is determined by activation of a hypothalamic gnrh pulse generator (for review, see refs.2 and 3). the pubertal increase in gnrh secretion is, in turn, prompted by changes in transsynaptic and glial inputs to the gnrh neuronal network. studies conducted by different groups have identified these inputs as being both facilitatory and inhibitory. the former use excitatory amino acids (for review, see refs.2 and 4) and the recently identified neuropeptide metastin/kisspeptin (5, 6) for neurotransmission/neuromodulation. -aminobutyric acid (gaba) and opioid peptides provide the inhibitory inputs (7, 8). it is also clear that the pubertal activation of gnrh secretion can no longer be considered as an event driven solely by transsynaptic inputs (2, 9). glial cells produce cell-cell signaling molecules that stimulate gnrh release and that have been shown to be critical for the correct timing of the pubertal process (for review, see ref.10).

    this article will briefly review our current understanding of the cell-cell mechanisms underlying the neuroendocrine control of puberty. we will also begin to develop the broad (but obviously imperfect) concept that the pubertal activation of gnrh secretion is controlled by a network of genes that, having diverse functions, operate within different cell contexts to coordinate the secretory activity of the gnrh neuronal network at puberty. we will propose the concept that coordination of gnrh release requires the participation of two sets of genes, those that are subordinate and those that govern the pubertal process at a higher hierarchical, transcriptional level of control.

    using a systems biology approach, i.e. the coordinated study of a biological system (11), for the understanding of these neuroendocrine regulatory networks, requires by definition (12): 1) identifying the genes, proteins, and other small molecules constituting the pathway of interest; 2) perturbing each pathway component through genetic manipulations and detecting the global cellular response to each perturbation with the help of high-throughput and whole-genomics techniques; 3) integrating the observed mrna and protein responses with existing models of protein-protein, protein-dna, and other interactions, using appropriate computational methods; and 4) formulating new hypotheses to explain observations not predicted by the model. as the reader will readily appreciate from this article, the application of these principles to the understanding of the neuroendocrine control of puberty is, at best, in an early embryonic stage.

    subordinate genes required for cell-cell communication

    this category include all downstream genes that participate in the excitatory and inhibitory control of gnrh neurons, whether this control is exerted transsynaptically or via glia-to-neuron communication (fig. 1). subordinate genes execute specific cellular functions required for cell-cell signaling, but their expression is hypothetically regulated by a higher order of system network control. for operational purposes they can be considered as the last to be activated.

    it was earlier established that the major excitatory transsynaptic event prompting the initiation of puberty is an increase in glutamatergic neurotransmission (2, 13), the primary mode of excitatory transsynaptic communication in the hypothalamus (14). activation of glutamatergic inputs increases gnrh secretion (15, 16) and accelerates sexual maturation in both rats and monkeys (17, 18). glutamate acts both directly (19, 20, 21, 22) and via regulatory neuronal subsets (23) to stimulate gnrh secretion.

    glutamatergic neurotransmission is a complex process controlled by a plethora of genes required for synthesis, transport, and release of the amino acid, as well as for the expression of the various receptors that mediate glutamate actions. puberty-related changes in glutamate receptor expression might be restricted to specific hypothalamic cellular subsets. for instance, the binding capacity of nmda and kainate receptors (which presumably reflect changes in gene expression) does not change in cell membranes derived from whole hypothalami (24). on the other hand, kainate receptor expression measured by in situ hybridization increases in gnrh neurons during sexual development (21). the upstream genes controlling this change are not known. even less is known about the transcriptional control of genes encoding enzymes involved in the synthesis, metabolism, and transport of glutamate. the importance of these homeostatic systems has been made evident by recent studies in which we used a quantitative proteomics approach (25) to identify proteins whose expression is increased in the hypothalamus at the time of rat puberty (26). we observed that the abundance of glutamate dehydrogenase, one of the enzymes that catalyzes the synthesis of glutamate (27), increases in the hypothalamus of female rats undergoing puberty. in contrast, the abundance of glutamine synthase, which catalyzes the metabolism of glutamate into glutamine (27), decreases at this time (fig. 1). these changes were accompanied by an increased capacity of the hypothalamus to release glutamate after blockade of glutamate transport, suggesting that more glutamate is available for both synaptic transmission and glia-to neuron signaling at the time of puberty. because both enzymes are predominantly expressed in glial cells, the results also indicate that an increased glutamate output of glial origin plays a major role in the control of gnrh release at puberty. the upstream genes controlling the transcriptional activity of the glutamate dehydrogenase and glutamine synthase genes remain to be identified. transcriptional regulation of vesicular glutamate transporter expression might represent an even more important control point because vesicular glutamate transporters (28, 29) are critical for the homeostatic minute-to-minute control of glutamate release (30).

    much discussion has been centered on the question of the primus movens of puberty: is it the loss of a central restraint (7) or the activation of stimulatory inputs to gnrh neurons (2) the recent finding that mutations of gpr54, the receptor for the kiss1-derived peptide metastin (31, 32, 33), causes hypothalamic hypogonadism (5, 6) suggests that the latter view is correct because gpr54 signaling is coupled to stimulation of gnrh release, instead of inhibition (34, 35). the kiss1-gpr54 signaling complex is a novel, and unsuspected, system involved in the control of gnrh secretion. metastin/kisspeptin is a 53-amino acid peptide encoded by the kiss1 gene (5, 6); proteolytic cleavage of the primary kiss1 protein product originates the decapeptide kisspeptin-10, which is extraordinarily potent in eliciting lh release (36, 37, 38). the comparable effectiveness of intracerebral and systemic administration (36, 39) suggests either a dual hypothalamic-pituitary site of action or a main effect at the median eminence (me) of the hypothalamus, a region of the brain located outside the blood-brain barrier. although kiss1-containing neurons are located in discrete neuronal subsets of the preoptic area (37) and the arcuate nucleus (36, 37), gpr54-containing cells are more diffusely distributed (36, 40), including gnrh neurons (40, 41) and the adenohypophysis (31, 32). this distribution suggests that kiss1 neurons may not only facilitate gnrh secretion by acting on gnrh neuronal perikarya and gnrh nerve terminals at the me but also stimulate gonadotropin secretion directly (39) by releasing metastin into the portal system. kiss1 and gpr54 mrna abundance increases in the nonhuman primate hypothalamus at the time of puberty, indicating that increased gpr54-mediated signaling contributes to the pubertal activation of gnrh secretion (36) (fig. 1). the ability of centrally administered kisspeptin to advance puberty in juvenile female rats (38, 39) supports this concept.

    gabaergic neurons acting via gabaa receptors provide a major inhibitory transsynaptic influence controlling gnrh secretion during prepubertal development (42, 43). although this restraining influence has been unambiguously demonstrated in primates (42, 44, 45), an inhibitory role of gabaergic neurotransmission in rodent puberty is much less clear because both inhibitory and stimulatory effects have been reported (for review, see ref.2). like glutamate, gaba regulates gnrh secretion by binding to receptors located both on gnrh neurons (46, 47) and on their synaptically connected neuronal partners (for review, see refs.2 and 4). it appears that the main effect of gaba acting via gabaa reception on gnrh neurons is excitation (47), but inhibitory effects have also been reported (46). like glutamate, gaba production requires the participation of different proteins involved in the synthesis, metabolism, transport, and release of the amino acid. because no changes in hypothalamic expression of the mrnas encoding gad-65 and gad-67 (the enzymes responsible for gaba synthesis) have been detected during primate sexual development (48), it does not appear that regulation of their gene expression is an event related to the onset of puberty. by analogy to the glutamatergic system, however, it might be inferred that important control points reside at the level of gaba vesicular transport (30, 49) and/or gaba receptors (fig. 1). again, the upstream genes involved in the transcriptional control of these components at the time of puberty remain to be identified.

    the other major inhibitory transsynaptic input to the gnrh neuronal network is provided by opiatergic neuronal systems (such as preproenkephalin-containing neurons). a reduction in opioid input to the gnrh neuronal network at the time of puberty may not be as critical as the loss of gabaergic inhibitory control. however, opioid peptides may provide additional homeostatic counter balance to the cascade of excitatory events leading to the pubertal increase in gnrh output. in rodents, the strength of the prepubertal opioid peptide inhibitory tone (50) diminishes at the time of puberty (51). opioid peptides as a group do not appear to restrain the initiation of puberty (for review, see refs.2 and 52), but it is possible that this inhibitory tone is exerted by a neuronal subset selectively using -endorphin, dynorphin, or met/leu-enkephalin for neurotransmission (fig. 1).

    gnrh neurons and glial cells share an intimate morphological association (53). in the me, both astroglia (53, 54, 55) and modified ependymoglial cells known as tanycytes (55, 56) appose gnrh terminals. tanycytic end-feet contacting the portal vessels intervene between gnrh nerve endings and endothelial cells of the portal vessels (55, 56) but retract at the time of the preovulatory surge of gonadotropins allowing the terminals to directly contact the endothelial cells (57). it is now clear that glial cells and gnrh neurons also share a functional relationship. this relationship depends upon growth factors acting via serine threonine kinase receptors, such as tgf1, and growth factors signaling through receptors with tyrosine kinase activity, like igf-i, basic fibroblast growth factor, and the members of the epidermal growth factor family, tgf, and neuregulins (nrgs) (fig. 1). we will discuss here only the latter because their role in the control of puberty has been more extensively characterized than the others. a more comprehensive discussion of the roles of igf-1 and basic fibroblast growth factor in the control of gnrh neurons can be found in (2, 58, 59).

    tgf binds to erbb1 receptors located on astrocytes and tanycytes, whereas nrgs are recognized by erbb4 receptors expressed only in astrocytes. both receptors recruit the coreceptor erbb2 for signaling, and in both cases, a major outcome is the release of chemical messengers, such as prostaglandin e2 (pge2), that act directly on gnrh neurons to stimulate gnrh secretion (60, 61, 62) (fig. 1). pharmacological and genetic approaches have been used to define the involvement of glial erbb1 receptors in the control of female sexual development. although blockade of erbb1 receptors in the me (63) or a point mutation of the erbb1 gene (64) result in delayed puberty, sexual development is advanced in transgenic mice conditionally overexpressing the tgf gene (65) and rats carrying intrahypothalamic grafts of cells genetically engineered to secrete tgf (66). ligand-dependent activation of erbb1 receptors in tanycytes results in plastic changes that, involving pge2 and tgf1 as downstream effectors, mimic the morphological plasticity displayed by tanycytes during the hours encompassing the preovulatory surge of gnrh (62). erbb1 signaling also has been implicated in the etiology of precocious puberty induced by hypothalamic hamartomas (hhs) in humans (67).

    in vivo disruption of hypothalamic erbb2 receptor synthesis using antisense oligodeoxynucleotides resulted in delayed puberty (61). such a delay was also observed in transgenic mice overexpressing, in an astrocyte-specific fashion, a truncated erbb4 protein (dnerbb4) that, lacking the intracellular domain, acts as a dominant negative receptor to block the signaling capability of the intact receptor (68). a combined deficiency achieved by generating dnerbb4 mice carrying a point mutation of the erbb1 receptors accentuated the effect of the single deficiencies (69), indicating that both systems work in a coordinated fashion to facilitate the onset of female puberty.

    are there mechanisms in place able to coordinate the transsynaptic and glial influences on gnrh neurons one of these mechanisms, initiated by excitatory amino acids, has been shown to target the astrocytic erbb signaling system for regulation (70). hypothalamic astrocytes express metabotropic and ionotropic -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. upon concomitant stimulation of both receptor subtypes, astrocytes respond with mobilization of erbb receptors to the cell surface and tgf/nrg-dependent phosphorylation of these receptors, indicating that glutamate stimulation of astrocytes facilitates the interaction of tgf/nrg ligands with their receptors (70). studies in other cell systems have shown that a surface protein with adhesion and protease activity termed tumor necrosis factor--converting enzyme or a disintegrin and metalloproteinase-17 cleaves tgf and nrgs from their transmembrane precursors, allowing the growth factors to bind their erbb receptors (71, 72). this is precisely what activation of -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and metabotropic glutamate receptors does in astrocytes, i.e. it enhances tumor necrosis factor--converting enzyme-like activity, which in turn elicits tgf release (73) (fig. 1).

    second tier genes controlling cell-cell interactions

    the late, but still present, initiation of puberty displayed by animals in which expression of certain candidate genes has been reduced (e.g. tgf, glutamate receptors, etc.) and the very low incidence in the human population of hypothalamic hypogonadism attributed to a single gene defect (for instance, gpr54) suggest that no isolated pathway or cellular subset is solely responsible for the neuroendocrine control of puberty. instead, this control is more likely exerted by functionally interconnected regulatory networks. as indicated in the introductory section, a first step in the implementation of a system biology approach is to identify all the genes, proteins, and other molecules constituting the pathway of interest (12). the conventional single gene/single protein approaches referred to in the previous section do obviously contribute to accomplish this first step. however, a thorough investigation of the diverse constituents of the various cellular networks implicated in the process requires the use of global, high-throughput approaches. using a combination of dna microarrays, proteomics, guilt by association, and retrospective approaches, we have singled out a group of genes that may represent a novel genetic network involved in the neuroendocrine control of female puberty (74). these genes have diverse cellular functions but share the common feature of having been earlier identified as involved in tumor suppression.

    with the help of dna microarrays, we queried the hypothalamus of female rhesus monkey at different phases of pubertal development and hhs from human subjects in search of candidate gene transcripts. we investigated hhs because these rare, nonneoplastic congenital malformations of the basal hypothalamus are usually associated with sexual precocity (75). to complement this genomic approach, we used quantitative proteomics (25) to identify hypothalamic proteins that might be down- or up-regulated in dnerbb4 mice, which as indicated above have delayed puberty (68). analysis of the monkey arrays results showed that expression of four tumor-related genes that otherwise participate in normal cell differentiation processes increases selectively in the hypothalamus at the time of monkey puberty (74). the hamartoma arrays identified four additional candidates also implicated in tumor pathology (76). in keeping with these observations, quantitative proteomics using isotope-coded affinity tag labeling revealed that the content of syncam, an immunoglobulin-like adhesion molecule required for synapse formation (77), was decreased in the dnerbb4 mice (78). before its synaptic function was discovered, syncam was known as tumor-suppressor in lung cancer-1 (79). quantitative pcr studies verified the array results (74, 76) and suggested that kiss1 and gpr54 are also part of this neuroendocrine gene network because their mrna expression increases in the hypothalamus at the time of primate puberty (36, 74). before the role of this complex in the control of puberty was discovered, the kiss1 gene was known as a suppressor of tumor metastases (33, 80). it is thus possible that syncam and kiss1 are members of a network of genes that, instead of functioning in the neuroendocrine brain to suppress tumor formation, serves to integrate neuron-to-neuron and glia-to-neuron communication into a functional unit capable of initiating the pubertal process (fig. 2). verification of this broad hypothesis will require implementation of the second step proposed for the application of a system biology strategy (12) to the understanding of the neuroendocrine control of puberty, that is, the controlled perturbation of specific genes followed by the global detection of the attendant cellular responses and the formulation, via repeated iterations, of specific models predicting the functional organization of the network. formidable challenges to be met are the elucidation of the developmental and spatial characteristics of the system.

    the upper echelon genes

    the concept that gnrh neuronal function and, hence, sexual development, is ultimately controlled by a hierarchy of upstream transcriptional regulators acting within functionally linked neuronal and glial subsets (81) is also in its infancy. an underlying premise is that, regardless of the transcriptional process they might control, the net outcome is to establish the conditions required for the productive interaction of neurons and glial cells (fig. 3). importantly, such genes not only reside at the core of any complex regulatory network (for instance, see refs.82 and 83), but they are also required to maintain the hierarchical structure of the network and to ensure that the system contains both redundancy and combinatorial diversity (83). thus far, we have identified three potential candidates for such a role. one of them is oct-2, a transcriptional regulator of the pou-domain family of homeobox-containing genes (84). although the oct-2 gene is expressed throughout the embryonic ventral forebrain, after birth its expression is restricted to a few hypothalamic neuronal subsets (85). oct-2 proteins are more abundant in cultured astrocytes than in neurons (86), suggesting that the oct-2 gene may be important for the trans-regulation of astroglial gene transcription. tgf has been identified as one of such targets (87), but the syncam promoter also contains oct-2 binding sites. the transcriptional activity of the tgf promoter is increased by oct-2. this regulatory mechanism is important for the onset of female puberty because: 1) hypothalamic oct-2 mrna levels increase during juvenile development in a gonad-independent manner, 2) blockade of oct-2 synthesis via antisense oligodeoxynucleotides reduced astrocytic tgf synthesis and delayed the age at first ovulation, and 3) lesions of the hypothalamus that induce sexual precocity activate both oct-2 and tgf expression in astrocytes near the lesion site (87).

    the second candidate is ttf-1 (thyroid transcription factor-1), another homeobox gene. ttf-1 is required for diencephalic morphogenesis (88); after birth it remains expressed in selected neuronal and glial populations of the hypothalamus, such as gnrh and preproenkephalinergic neurons and tanycytes of the me (89). ttf-1 acts on each of these cell types to promote cell-specific functions. for instance, it enhances gnrh and erbb2 gene transcription but inhibits preproenkephalin promoter activity (89). dna arrays and quantitative pcr analysis of the female rhesus monkey hypothalamus revealed a pubertal increase in ttf-1 expression. employing the cre-loxp system to conditionally delete the ttf-1 gene from those neuronal subsets of the hypothalamus where it is normally expressed, we found that ttf-1 null mutants have delayed puberty, a disruption of initial estrous cyclicity, and decreased reproductive capacity (90). these deficiencies were accompanied by increased preproenkephalin gene expression (90) and by suppressed hypothalamic gnrh and kiss1 mrna levels (mastronardi, c. a., g. smiley, t. kusakabe, a. kawagushi, s. heger, r. cabrera, a. e. mungenast, s. kimura, and s. r. ojeda, unpublished data). thus, ttf1 enhances the transcriptional activity of genes required for the facilitatory control of puberty (gnrh, erbb2, kiss1) while repressing the transcription of a gene involved in the inhibition of gnrh secretion.

    the third candidate was also discovered using cdna arrays to interrogate the primate hypothalamus at the time of puberty (91). it is a gene earlier known as c14orf4 (92), but that we have now termed eap-1 (91). like ttf-1, eap-1 maps to human chromosome 14. hypothalamic eap-1 mrna levels increase in both monkeys and rats during female puberty (93), suggesting an involvement in the control of the pubertal process. eap-1 encodes a nuclear protein, which is expressed in neuronal subsets involved in the stimulatory and inhibitory control of gnrh secretion, such as glutamatergic, gabaergic, proenkephalinergic, and kiss1 neurons, in addition to gnrh neurons themselves (93). like ttf-1, eap-1 transactivates the promoter of genes involved in facilitating the advent of puberty (e.g. gnrh) while suppressing the expression of genes inhibitory to the pubertal process (such as the preproenkephalin gene). knocking down hypothalamic eap-1 expression via sirna technology delayed puberty and disrupted estrous cyclicity, confirming the importance of eap-1 as an upper echelon gene necessary for the neuron-to neuron regulation of gnrh secretion at puberty (93) (fig. 3).

    conclusion

    these observations suggest that the neuroendocrine control of puberty is provided by a gene network of hierarchical nature similar in principle to those postulated to exist in less complex cellular systems (see refs.12 , 82 , and 83 and references therein). essential features of such networks are the dominance of a few highly connected upper echelon gene hubs, the partial overlap of second tier gene subnetworks, the large number of less-connected subordinate gene nodes, and the remarkable redundancy of the system (83). although the overall validity of this concept (summarized in figs. 2 and 3) remains to be experimentally tested, the existence of a hypothalamic gene network composed of genes situated at different, but interactive, hierarchical levels is consistent with the idea that the onset of puberty is genetically determined and depends on the contribution of more than one gene (94, 95, 96). further supporting this idea is the recent identification of key quantitative trait loci regulating the abundance of thousands of transcripts in the nervous system in a region-specific manner (97). recent reports have also made clear that, contradicting the current dogma that human central precocious puberty is sporadic in nature, a significant number of cases of this disorder is caused by genetic factors (98). future studies should make clear whether those genes implicated in the control of puberty in animal models are, in fact, required for the normalcy of human puberty.

    footnotes

    this work was supported in part by national institutes of health grants hd25123, mh65438, hd050798, and rr00163 and through cooperative agreement u-54 hd18185 as part of the specialized cooperative center’s program in reproduction research.

    present address for c.r.: department of pediatrics, university of bonn, adenauerallee 119, 53113 bonn, germany.

    present address for a.e.m.: department of neuroscience, school of medicine, university of pennsylvania, philadelphia, pennsylvania 19104.

    present address for a.-s.p.: departement des sciences cliniques, universite de liege, liege 4000, belgium.

    a.l., c.m., s.h., c.r., a.s.p., v.m., and a.e.m. have nothing to declare. s.r.o. received a lecture fee from ferring co. (berkshire, uk).

    first published online december 22, 2005

    abbreviations: gaba, -aminobutyric acid; hh, hypothalamic hamartoma; me, median eminence; nrg, neuregulin; pg, prostaglandin; tsg, tumor suppressor gene; ttf-1, thyroid transcription factor-1.

    accepted for publication november 21, 2005.

    references

    boyar r, finkelstein j, roffwarg h, kapen s, weitzman e, hellman l 1972 synchronization of augmented luteinizing hormone secretion with sleep during puberty. n engl j med 287:582586

    ojeda sr, terasawa e 2002 neuroendocrine regulation of puberty. in: pfaff d, arnold a, etgen a, fahrbach s, moss r, rubin r, eds. hormones, brain and behavior. vol 4. new york: elsevier; 589659

    plant tm 2002 neurophysiology of puberty. j adolesc health 31:185191

    ojeda sr, skinner mk 2005 puberty in the rat. in: neill jd, ed. the physiology of reproduction. 3rd ed. san diego: academic press/elsevier; 20612126

    de roux n, genin e, carel j-c, matsuda f, chaussain j-l, milgrom e 2003 hypogonadotropic hypogonadism due to loss of function of the kiss1-derived peptide receptor gpr54. proc natl acad sci usa 100:1097210976

    seminara sb, messager s, chatzidaki ee, thresher rr, acierno jr js, shagoury jk, bo-abbas y, kuohung w, schwinof km, hendrick ag, zahn d, dixon j, kaiser ub, slaugenhaupt sa, gusella jf, o’rahilly s, carlton mb, crowley jr wf, aparicio sa, colledge wh 2003 the gpr54 gene as a regulator of puberty. n engl j med 349:16141627

    terasawa e, fernandez dl 2001 neurobiological mechanisms of the onset of puberty in primates. endocr rev 22:111151

    terasawa e 1999 hypothalamic control of the onset of puberty. curr opin endocrinol diabetes 6:4449

    ojeda sr, ma yj, dziedzic b, prevot v 2000 astrocyte-neuron signaling and the onset of female puberty. in: bourguignon j-p, plant tm, eds. the onset of puberty in perspective. amsterdam: elsevier science b.v.; 4157

    ojeda sr, prevot v, heger s, lomniczi a, dziedzic b, mungenast a 2003 glia-to neuron signaling and the neuroendocrine control of female puberty. ann med 35:244255

    klipp e, herwig r, kowald a, wierling c, lehrach h 2005 systems biology in practice. weinheim, germany: wiley-vch; 3449

    ideker t, thorsson v, ranish ja, christmas r, buhler j, eng jk, bumgarner r, goodlett dr, aebersold r, hood l 2001 integrated genomic and proteomic analyses of a systematically perturbed metabolic network. science 292:929934

    bourguignon j-p, lebrethon mc, gerard a, purnell g, vandersmissen e, parent as, yamanaka c 2000 amino acid neurotransmission and early ontogeny of pulsatile gnrh secretion from the rat hypothalamus. in: bourguignon j-p, plant tm, eds. the onset of puberty in perspective. amsterdam: elsevier science b.v.; 119129

    van den pol an, trombley pq 1993 glutamate neurons in hypothalamus regulate excitatory transmission. j neurosci 13:28292836

    claypool le, kasuya e, saitoh y, marzban f, terasawa e 2000 n-methyl d,l-aspartate induces the release of luteinizing hormone-releasing hormone in the prepubertal and pubertal female rhesus monkey as measured by in vivo push-pull perfusion in the stalk-median eminence. endocrinology 141:219228

    donoso ao, lopez fj, negro-vilar a 1990 glutamate receptors of the non-n-methyl-d-aspartic acid type mediate the increase in luteinizing hormone releasing hormone release by excitatory amino acid in vitro. endocrinology 126:414420

    plant tm, gay vl, marshall gr, arslan m 1989 puberty in monkeys is triggered by chemical stimulation of the hypothalamus. proc natl acad sci usa 86:25062510

    urbanski hf, ojeda sr 1990 a role for n-methyl-d-aspartate (nmda) receptors in the control of lh secretion and initiation of female puberty. endocrinology 126:17741776

    ottem en, godwin jg, petersen sl 2002 glutamatergic signaling through the n-methyl-d-aspartate receptor directly activates medial subpopulations of luteinizing hormone-releasing hormone (lhrh) neurons, but does not appear to mediate the effects of estradiol on lhrh gene expression. endocrinology 143:48374845

    gore ac 2001 gonadotropin-releasing hormone neurons. nmda receptors, and their regulation by steroid hormones across the reproductive life cycle. brain res rev 37:235248

    eyigor o, jennes l 1997 expression of glutamate receptor subunit mrnas in gonadotropin-releasing hormone neurons during the sexual maturation of the female rat. neuroendocrinology 66:122129

    eyigor o, jennes l 2000 kainate receptor subunit-positive gonadotropin-releasing hormone neurons express c-fos during the steroid-induced luteinizing hormone surge in the female rat. endocrinology 141:779786

    van den pol an, wuarin j-p, dudek fe 1990 glutamate, the dominant excitatory transmitter in neuroendocrine regulation. science 250:12761278

    brann dw, zamorano pl, ping l, mahesh vb 1993 role of excitatory amino acid neurotransmission during puberty in the female rat. mol cell neurosci 4:107112

    gygi sp, rist b, gerber sa, turecek f, gelb mh, aebersold r 1999 quantitative analysis of complex protein mixtures using isotope-coded affinity tags. nat biotechnol 17:994999

    roth cl, mccormack al, lomniczi a, mungenast ae, ojeda sr, quantitative proteomics identifies a major change in glial glutamate metabolism at the time of female puberty. mol cell endocrinol, in press

    erecinska m, silver ia 1990 metabolism and role of glutamate in mammalian brain. prog neurobiol 35:245296

    takamori s, rhee js, rosenmund c, john r 2000 identification of a vesicular glutamate transporter that defines a glutamatergic phenotype in neurons. nature 407:189194

    fremeau jr rt, troyer md, pahner i, nygaard go, tran ch, reimer rj, bellocchio ee, fortin d, storm-mathisen j, edwards rh 2001 the expression of vesicular glutamate transporters defines two classes of excitatory synapse. neuron 31:247260

    de gois s, schafer mk, defamie n, chen c, ricci a, weihe e, varoqui h, erickson jd 2005 homeostatic scaling of vesicular glutamate and gaba transporter expression in rat neocortical circuits. j neurosci 25:71217133

    muir ai, chamberlain l, elshourbagy na, michalovich d, moore dj, calamari a, szekeres pg, sarau hm, chambers jk, murdock p, steplewski k, shabon u, miller je, middleton se, darker jg, larminie cg, wilson s, bergsma dj, emson p, faull r, philpott kl, harrison dc 2001 axor12, a novel human g protein-coupled receptor, activated by the peptide kiss-1. j biol chem 276:2896928975

    kotani m, detheux m, vandenbogaerde a, communi d, vanderwinden jm, le poul e, brezillon s, tyldesley r, suarez-huerta n, vandeput f, blanpain c, schiffmann sn, vassart g, parmentier m 2001 the metastasis suppressor gene kiss-1 encodes kisspeptins, the natural ligands of the orphan g protein-coupled receptor gpr54. j biol chem 276:3463134636

    ohtaki t, shintani y, honda s, matsumoto h, hori a, kanehashi k, terao y, kumano s, takatsu y, masuda y, ishibashi y, watanabe t, asada m, yamada t, suenaga m, kitada c, usuki s, kurokawa t, onda h, nishimura o, fujino m 2001 metastasis suppressor gene kiss-1 encodes peptide ligand of a g-protein-coupled receptor. nature 411:613617

    navarro vm, castellano jm, fernández-fernández r, barreiro ml, roa j, sanchez-criado je, aguilar e, dieguez c, pinilla l, tena-sempere m 2004 developmental and hormonally regulated messenger ribonucleic acid expression of kiss-1 and its putative receptor, gpr54, in rat hypothalamus and potent luteinizing hormone-releasing activity of kiss-1 peptide. endocrinology 145:45654574

    shahab m, mastronardi c, plant tm, ojeda sr, crowley jr wf, seminara sb, hypothalamic gpr-54 expression and signaling during the peripubertal period in the rhesus monkey (macaca mulatta). program of the 86th annual meeting of the endocrine society, new orleans, la, 2004, p 529 (abstract p3-269)

    shahab m, mastronardi c, seminara sb, crowley wf, ojeda sr, plant tm 2005 increased hypothalamic gpr54 signaling: a potential mechanism for initiation of puberty in primates. proc natl acad sci usa 102:21292134

    gottsch ml, cunningham mj, smith jt, popa sm, acohido bv, crowley wf, seminara s, clifton dk, steiner ra 2004 a role for kisspeptins in the regulation of gonadotropin secretion in the mouse. endocrinology 145:40734077

    navarro vm, fernandez-fernandez r, castellano jm, roa j, mayen a, barreiro ml, gaytan f, aguilar e, pinilla l, dieguez c, tena-sempere m 2004 advanced vaginal opening and precocious activation of the reproductive axis by kiss-1 peptide, the endogenous ligand of gpr54. j physiol 561:379386

    navarro vm, castellano jm, fernández-fernández r, tovar s, roa j, mayen a, nogueiras r, vazquez mj, barreiro ml, magni p, aguilar e, dieguez c, pinilla l, tena-sempere m 2005 characterization of the potent luteinizing hormone-releasing activity of kiss-1 peptide, the natural ligand of gpr54. endocrinology 146:156163

    irwig ms, fraley gs, smith jt, acohido bv, popa sm, cunningham mj, gottsch ml, clifton dk, steiner ra 2005 kisspeptin activation of gonadotropin releasing hormone neurons and regulation of kiss-1 mrna in the male rat. neuroendocrinology 80:264272

    parhar is, ogawa s, sakuma y 2004 laser captured single digoxigenin-labeled neurons of gonadotropin-releasing hormone types reveal a novel g protein-coupled receptor (gpr54) during maturation in cichlid fish. endocrinology 145:36133618

    mitsushima d, hei dl, terasawa e 1994 -aminobutyric acid is an inhibitory neurotransmitter restricting the release of luteinizing hormone-releasing hormone before the onset of puberty. proc natl acad sci usa 91:395399

    mitsushima d, kimura f 1997 the maturation of gabaa receptor-mediated control of luteinizing hormone secretion in immature male rats. brain res 748:258262

    mitsushima d, marzban f, luchansky ll, bruich aj, keen kl, durning m, golos tg, terasawa e 1996 role of glutamic acid decarboxylase in the prepubertal inhibition of the luteinizing hormone releasing hormone release in female rhesus monkeys. j neurosci 16:25632573

    keen kl, burich aj, mitsushima d, kasuya e, terasawa e 1999 effects of pulsatile infusion of the gabaa receptor blocker bicuculline on the onset of puberty in female rhesus monkeys. endocrinology 140:52575266

    han sk, todman mg, herbison ae 2004 endogenous gaba release inhibits the firing of adult gonadotropin-releasing neurons. endocrinology 145:495499

    defazio ra, heger s, ojeda sr, moenter sm 2002 activation of a-type -aminobutyric acid receptors excites gonadotropin-releasing hormone neurons. mol endocrinol 16:28722891

    urbanski hf, rodrigues sm, garyfallou vt, kohama sg 1998 regional distribution of glutamic acid decarboxylase (gad65 and gad67) mrna in the hypothalamus of male rhesus macaques before and after puberty. mol brain res 57:8691

    mcintire sl, reimer rj, schuske k, edwards rh, jorgensen em 1997 identification and characterization of the vesicular gaba transporter. nature 389:870876

    blank ms, panerai ae, friesen hg 1979 opioid peptides modulate hormone secretion during sexual development. science 203:11291131

    wilkinson m, bhanot r 1982 a puberty-related attenuation of opiate peptide-induced inhibition of lh secretion. endocrinology 110:10461048

    ojeda sr, urbanski hf 1994 puberty in the rat. in: knobil e, neill jd, eds. the physiology of reproduction. 2nd ed. vol 2. new york: raven press; 363409

    silverman a-j, livne i, witkin jw 1994 the gonadotropin-releasing hormone (gnrh), neuronal systems: immunocytochemistry and in situ hybridization. in: knobil e, neill jd, eds. the physiology of reproduction. 2nd ed. vol 1. new york: raven press; 16831709

    witkin jw, ferin m, popilskis sj, silverman a-j 1991 effects of gonadal steroids on the ultrastructure of gnrh neurons in the rhesus monkey: synaptic input and glial apposition. endocrinology 129:10831092

    king jc, letourneau rl 1994 luteinizing hormone-releasing hormone terminals in the median eminence of rats undergo dramatic changes after gonadectomy, as revealed by electron microscopic image analysis. endocrinology 134:13401351

    kozlowski gp, coates pw 1985 ependymoneuronal specializations between lhrh fibers and cells of the cerebroventricular system. cell tissue res 242:301311

    king jc, rubin bs 1996 recruitment of lhrh neurons and increased access of lhrh terminals to portal capillaries: integral mechanisms for lh surge induction. ann endocrinol (paris) 57(suppl 4):72

    garcia-segura lm, mccarthy mm 2004 minireview: role of glia in neuroendocrine function. endocrinology 145:10821086

    gill jc, moenter sm, tsai p-s 2004 developmental regulation of gonadotropin-releasing hormone neurons by fibroblast growth factor signaling. endocrinology 145:38303839

    ma yj, berg-von der emde k, rage f, wetsel wc, ojeda sr 1997 hypothalamic astrocytes respond to transforming growth factor  with secretion of neuroactive substances that stimulate the release of luteinizing hormone-releasing hormone. endocrinology 138:1925

    ma yj, hill df, creswick ke, costa me, ojeda sr 1999 neuregulins signaling via a glial erbb2/erbb4 receptor complex contribute to the neuroendocrine control of mammalian sexual development. j neurosci 19:99139927

    prevot v, cornea a, mungenast a, smiley g, ojeda sr 2003 activation of erbb-1 signaling in tanycytes of the median eminence stimulates transforming growth factor b1 release via prostaglandin e2 production and induces cell plasticity. j neurosci 23:1062210632

    ma yj, junier m-p, costa me, ojeda sr 1992 transforming growth factor  (tgf) gene expression in the hypothalamus is developmentally regulated and linked to sexual maturation. neuron 9:657670

    apostolakis em, garai j, lohmann je, clark jh, o’malley bw 2000 epidermal growth factor activates reproductive behavior independent of ovarian steroids in female rodents. mol endocrinol 14:10861098

    ma yj, dissen ga, merlino g, coquelin a, ojeda sr 1994 overexpression of a human transforming growth factor alpha (tgf) transgene reveals a dual antagonistic role of tgf in female sexual development. endocrinology 135:13921400

    rage f, hill df, sena-esteves m, breakefield xo, coffey rj, costa me, mccann sm, ojeda sr 1997 targeting transforming growth factor  expression to discrete loci of the neuroendocrine brain induces female sexual precocity. proc natl acad sci usa 94:27352740

    jung h, carmel p, schwartz ms, witkin jw, bentele khp, westphal m, piatt jh, costa me, cornea a, ma yj, ojeda sr 1999 some hypothalamic hamartomas contain transforming growth factor , a puberty-inducing growth factor, but not luteinizing hormone-releasing hormone neurons. j clin endocrinol metab 84:46954701

    prevot v, rio c, cho gj, lomniczi a, heger s, neville cm, rosenthal na, ojeda sr, corfas g 2003 normal female sexual development requires neuregulin-erbb receptor signaling in hypothalamic astrocytes. j neurosci 23:230239

    prevot v, lomniczi a, corfas g, ojeda sr 2005 erbb-1 and erbb-4 receptors act in concert to facilitate both female sexual development and mature reproductive function. endocrinology 146:14651472

    dziedzic b, prevot v, lomniczi a, jung h, cornea a, ojeda sr 2003 neuron-to-glia signaling mediated by excitatory amino acid receptors regulates erbb receptor function in astroglial cells of the neuroendocrine brain. j neurosci 23:915926

    peschon jj, slack jl, reddy p, stocking kl, sunnarborg sw, lee dc, russell we, castner bj, johnson rs, fitzner jn, boyce rw, nelson n, koslosky cj, wolfson mf, rauch ct, cerretti dp, paxton rj, march cj, black ra 1998 an essential role for ectodomain shedding in mammalian development. science 282:12811284

    sahin u, weskamp g, kelly k, zhou h-m, higashiyama s, peshon j, hartmann d, saftig p, blobel cp 2004 distinct roles for adam10 and adam17 in ectodomain shedding of six egfr ligands. j cell biol 164:769779

    lomniczi a, cornea a, costa me, ojeda sr, hypothalamic tumor necrosis factor- converting enzyme (tace) mediates excitatory amino acid-dependent neuron-to-glia signaling in the neuroendocrine brain. j neurosci, in press

    roth cl, mastronardi c, mungenast a, heger s, jung h, ojeda sr 2004 gene expression profiling of the nonhuman primate hypothalamus at the time of female puberty reveals activation of tumor suppressor gene expression. horm res 62(suppl 2):pl-69

    grumbach mm, styne dm 2003 puberty: ontogeny, neuroendocrinology, physiology, and disorders. in: larsen pr, kronenberg hm, melmed s, polonsky ks, eds. williams textbook of endocrinology. 10th ed. philadelphia: w.b. saunders; 11151286

    parent as, jung h, westphal m, ojeda sr, gene expression profiling of hypothalamic hamartomas: a search for genes involved in initiating human puberty. program of the 87th annual meeting of the endocrine society, san francisco, 2005, p 190 (abstract p1-94)

    biederer t, sara y, mozhayeva m, atasoy d, liu x, kavalali et, südhof tc 2002 syncam, a synaptic adhesion molecule that drives synapse assembly. science 297:15251531

    mungenast ae, parent a, chen ss, goodlett d, aebersold r, corfas g, ojeda sr 2003 the synaptic adhesion molecule syncam is associated with erbb4 dysregulation in the hypothalamus of mice with a delayed onset of puberty. program no 281 20, 2003 abstract viewer washington, dc: society for neuroscience, 2003 online

    kuramochi m, fukuhara h, nobukuni t, kanbe t, maruyama t, ghosh hp, pletcher m, isomura m, onizuka m, kitamura t, sekiya t, reeves rh, murakami y 2001 tslc1 is a tumor-suppressor gene in human non-small-cell lung cancer. nat genet 27:427430

    steeg ps, ouatas t, halverson d, palmieri d, salerno m 2003 metastasis suppressor genes: basic biology and potential clinical use. clin breast cancer 4:5162

    ojeda sr 1991 the mystery of mammalian puberty: how much more do we know perspect biol med 34:365383

    davidson eh, rast jp, oliveri p, ransick a, calestani c, yuh c-h, minokawa t, amore g, hinman v, arenas-mena c, otim o, brown ct, livi cb, lee py, revilla r, rust ag, pan zj, schilstra mj, clarke pjc, arnone mi, rowen l, cameron ra, mcclay dr, hood l, bolouri h 2002 a genomic regulatory network for development. science 295:16691678

    basso k, margolin aa, stolovitzky g, klein u, dalla-favera r, califano a 2005 reverse engineering of regulatory networks in human b cells. nat genet 37:382390

    treacy mn, rosenfeld mg 1992 expression of a family of pou-domain protein regulatory genes during development of the central nervous system. annu rev neurosci 15:139165

    alvarez-bolado g, rosenfeld mg, swanson lw 1995 model of forebrain regionalization based on spatiotemporal patterns of pou-iii homeobox gene expression, birthdates, and morphological features. j comp neurol 355:237295

    hatzopoulos ak, stoykova as, erselius jr, goulding m, neuman t, gruss p 1990 structure and expression of the mouse oct2a and oct2b, two differentially spliced products of the same gene. development 109:349362

    ojeda sr, hill j, hill df, costa me, tapia v, cornea a, ma yj 1999 the oct-2 pou-domain gene in the neuroendocrine brain: a transcriptional regulator of mammalian puberty. endocrinology 140:37743789

    kimura s, hara y, pineau t, fernandez-salguero p, fox ch, ward jm, gonzalez fj 1996 the t/ebp null mouse: thyroid-specific enhancer-binding protein is essential for the organogenesis of the thyroid, lung, ventral forebrain, and pituitary. genes dev 10:6069

    lee bj, cho gj, norgren r, junier m-p, hill df, tapia v, costa me, ojeda sr 2001 ttf-1, a homeodomain gene required for diencephalic morphogenesis, is postnatally expressed in the neuroendocrine brain in a developmentally regulated and cell-specific fashion. mol cell neurosci 17:107126

    mastronardi c, smiley g, kuswakabe t, kawagushi a, cabrera r, mungenast a, kimura s, ojeda sr 2004 neuronal deletion of the t/ebp gene delays female puberty and causes premature reproductive senescence. program no 143 2, 2004 abstract viewer washington, dc: society for neuroscience, 2004 online

    ojeda sr, lomniczi a, mungenast a, mastronardi c, parent as, roth c, prevot v, heger s, jung h 2005 towards understanding the neurobiology of mammalian puberty: genetic, genomic and proteomic approaches. in: kordon c, gaillard r, christen y, eds. hormones and the brain. berlin: springer verlag; 4760

    rampazzo a, pivotto f, occhi g, tiso n, bortoluzzi s, rowen l, hood l, nava a, danieli ga 2000 characterization of c14orf4, a novel intronless human gene containing a polyglutamine repeat, mapped to the arvd1 critical region. biochem biophys res commun 278:766774

    heger s, mastronardi c, lomniczi a, cabrera r, roth c, sippell wg, jung h, dissen ga, ojeda sr 2005 role of a novel gene (enhanced at puberty, eap-1) in the regulation of female puberty. horm res 64(suppl 1):22

    krewson td, supelak pj, hill ae, singer jb, lander es, nadeau jh, palmert mr 2004 chromosomes 6 and 13 harbor genes that regulate pubertal timing in mouse chromosome substitution strains. endocrinology 145:44474451

    seminara sb, crowley jr wf 2001 perspective: the importance of genetic defects in humans in elucidating the complexities of the hypothalamic-pituitary-gonadal axis. endocrinology 142:21732177

    eaves l, silberg j, foley d, bulik c, maes h, erkanli a, angold a, costello ej, worthman c 2004 genetic and environmental influences on the relative timing of pubertal change. twin res 7:471481

    chesler ej, lu l, shou s, qu y, gu j, wang j, hsu hc, mountz jd, baldwin ne, langston ma, threadgill dw, manly kf, williams rw 2005 complex trait analysis of gene expression uncovers polygenic and pleiotropic networks that modulate nervous system function. nat genet 37:233242

    de vries l, kauschansky a, shohat m, phillip m 2004 familial central precocious puberty suggests autosomal dominant inheritance. j clin endocrinol metab 89:17941800

  • 上一个医药学论文:
  • 下一个医药学论文:
  •  作者:11665 [标签: ]
    姓 名: *
    E-mail:
    评 分: 1分 2分 3分 4分 5分
    评论内容:
    发表评论请遵守中国各项有关法律法规,评论内容只代表网友个人观点,与本网站立场无关。
    浅析模拟信号数字化研究及Simulink仿真技术
    Identity Construction in Eastern and Wes
    Seminar教学法在临床药理学课程中的应用
    汉黄芩素对胶质瘤U251细胞Survivin mRNA及C
    基于CMM/CMMI的SQA过程在信息系统开发中的应
    脑梗死患者血清NO及tNOS\iNOS的测定
    UF-1000i尿沉渣分析仪参数RBC-INFORMATION值
    浅谈药学研究生培养过程中实验室学术例会(L
    cyclinD1 P16 P27联合判断癌症患者病情的研
    E cadherin在胆管癌组织中的表达及意义
    Paxillin和Survivin在结肠癌中的表达及其意
    重组人生长激素对脑缺血 再灌注损伤细胞凋亡
    | 设为首页 | 加入收藏 | 联系我们 | 网站地图 | 手机版 | 论文发表

    Copyright 2006-2013 © 毕业论文网 All rights reserved 

     [中国免费论文网]  版权所有