151
|
Carlson NE, Horton KW, Grunwald GK. A comparison of methods for analyzing time series of pulsatile hormone data. Stat Med 2013; 32:4624-38. [PMID: 23787487 DOI: 10.1002/sim.5882] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 05/27/2013] [Indexed: 11/06/2022]
Abstract
Many endocrine systems are regulated by pulsatile hormones - hormones that are secreted intermittently in boluses rather than continuously over time. To study pulsatile secretion, blood is drawn every few minutes for an extended period. The result is a time series of hormone concentrations for each individual. The goal is to estimate pulsatile hormone secretion features such as frequency, location, duration, and amount of pulsatile and non-pulsatile secretion and compare these features between groups. Various statistical approaches to analyzing these data have been proposed, but validation has generally focused on one hormone. Thus, we lack a broad understanding of each method's performance. By using simulated data with features seen in reproductive and stress hormones, we investigated the performance of three recently developed statistical approaches for analyzing pulsatile hormone data and compared them to a frequently used deconvolution approach. We found that methods incorporating a changing baseline modeled both constant and changing baseline shapes well; however, the added model flexibility resulted in a slight increase in bias in other model parameters. When pulses were well defined and baseline constant, Bayesian approaches performed similar to the existing deconvolution method. The increase in computation time of Bayesian approaches offered improved estimation and more accurate quantification of estimation variation in situations where pulse locations were not clearly identifiable. Within the class of deconvolution models for fitting pulsatile hormone data, the Bayesian approach with a changing baseline offered adequate results over the widest range of data.
Collapse
Affiliation(s)
- N E Carlson
- Department of Biostatistics and Informatics, University of Colorado Denver, 13001 E. 17th PL, MS B119, Aurora, CO, 80047, U.S.A
| | | | | |
Collapse
|
152
|
|
153
|
Rick FG, Block NL, Schally AV. An update on the use of degarelix in the treatment of advanced hormone-dependent prostate cancer. Onco Targets Ther 2013; 6:391-402. [PMID: 23620672 PMCID: PMC3633549 DOI: 10.2147/ott.s32426] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Androgen deprivation therapy remains the mainstay of medical treatment for advanced prostate cancer. Commonly, this is achieved with medical androgen deprivation rather than surgical intervention as the permanence and psychological effects of the latter are unacceptable for most patients. Degarelix is a third generation antagonist of luteinizing hormone-releasing hormone (LHRH, also termed gonadotropin-releasing hormone) for the first-line treatment of androgen-dependent advanced prostate cancer. Degarelix acts directly on the pituitary receptors for LHRH, blocking the action of endogenous LHRH. The use of degarelix eliminates the initial undesirable surge in gonadotropin and testosterone levels, which is produced by agonists of LHRH. Degarelix is the most comprehensively studied and widely available LHRH antagonist worldwide. Clinical trials have demonstrated that degarelix has a long-term efficacy similar to the LHRH agonist leuprolide in achieving testosterone suppression in patients with prostate cancer. Degarelix, however, produces a faster suppression of testosterone and prostate-specific antigen (PSA), with no testosterone surges or microsurges, and thus prevents the risk of clinical flare in advanced disease. Recent clinical trials demonstrated that treatment with degarelix results in improved disease control when compared with an LHRH agonist in terms of superior PSA progression-free survival, suggesting that degarelix likely delays progression to castration-resistant disease and has a more significant impact on bone serum alkaline phosphatase and follicle-stimulating hormone. Degarelix is usually well tolerated, with limited toxicity and no evidence of systemic allergic reactions in clinical studies. Degarelix thus represents an important addition to the hormonal armamentarium for therapy of advanced androgen-dependent prostate cancer.
Collapse
Affiliation(s)
- Ferenc G Rick
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | | |
Collapse
|
154
|
Meczekalski B, Podfigurna-Stopa A, Smolarczyk R, Katulski K, Genazzani AR. Kallmann syndrome in women: from genes to diagnosis and treatment. Gynecol Endocrinol 2013; 29:296-300. [PMID: 23368665 DOI: 10.3109/09513590.2012.752459] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Kallmann syndrome (KS) can be characterized as genetic disorder marked by hypogonadotropic hypogonadism and anosmia. Franz Jozef Kallmann was the first who described this disease in 1944. He suggested, that this disease has hereditary background. At present, six genes are regarded as causal genes of KS. These genes can be listed in chronological order: KAL1, FGFR1, FGF8, CHD7, PROKR2 and PROK2. The sensitivity of molecular testing of KS is only about 30%. Diagnosis based on clinical findings is therefore such important. Cardinal features of patients with KS include hypogonadotropic hypogonadism and anosmia or hyposmia. Some non-reproductive, non-olfactory symptoms can also be present, depending on the genetic form of disease. Some patients with KS present midline cranial anomalies (cleft lip, cleft palate and imperfect fusion). Sometimes patients can also suffer from missing teeth (dental agenesis). Optic problems, such as colour blindness or optic atrophy also can occur in KS patients. Very characteristic symptom in KS patients is mirror movements of the upper limbs (imitation synkinesis for contralateral limbs). The type of treatment in women with KS depends on the goal of therapy. After the diagnosis of syndrome, the main goal of the treatment is to induce and maintain secondary sex characteristic (estrogen-progestin therapy). The further goal in some patients can be related to enable fertility (gonadotropin, gonadotropin-releasing hormone therapy).
Collapse
Affiliation(s)
- Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, Poznan, Poland.
| | | | | | | | | |
Collapse
|
155
|
Systems approaches to genomic and epigenetic inter-regulation of peptide hormones in stress and reproduction. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2013; 113:375-86. [PMID: 23500148 DOI: 10.1016/j.pbiomolbio.2013.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 02/08/2013] [Accepted: 02/21/2013] [Indexed: 12/20/2022]
Abstract
The evolution of the organismal stress response and fertility are two of the most important aspects that drive the fitness of a species. However, the integrated regulation of the hypothalamic pituitary adrenal (HPA) and hypothalamic-pituitary-gonadal (HPG) axes has been traditionally thwarted by the complexity of these systems. Pepidergic signalling systems have emerged as critical integrating systems for stress and reproduction. Current high throughput systems approaches are now providing a detailed understanding of peptide signalling in stress and reproduction. These approaches were dependent upon a long history of discovery aimed at the structural characterization of the associated molecular components. The combination of comparative genomics, microarray and epigenetic studies has led not only to a much greater understanding of the integration of stress and reproduction but also to the discovery of novel physiological systems. Recent epigenomic approaches have similarly yielded a new level of complexity in the interaction of these physiological systems. Together, such studies have provided a greater understanding of the effects of stress and reproduction.
Collapse
|
156
|
Abstract
In 1946, at the end of World War II, I entered graduate school at Cornell University, where I remained for 44 years. During that time, my laboratory produced more than 300 publications in the field of reproductive biology, including studies on nutrition and reproduction, the role of the hypothalamus in pituitary gonadotropin release, corpus luteum formation and function, hormone assays, and estrous cycle synchronization. At age seventy, I retired from Cornell and accepted the Gordon Cain Endowed Professorship at Louisiana State University, where I continued my work on the bovine corpus luteum and added research on the collection, maturation, in vitro fertilization, and culture of bovine oocytes. In 1994, I moved to the Pennington Biomedical Research Center and soon thereafter started the research that led to development of the lytic peptide-gonadotropin conjugates, which target and destroy cancer cell membranes. I am continuing my work on the development of targeted cancer cell drugs and, yes, research is still fun!
Collapse
Affiliation(s)
- William Hansel
- Liberty Hyde Bailey Emeritus Professor, Cornell University, Ithaca, New York 14853
| |
Collapse
|
157
|
Young J, George JT, Tello JA, Francou B, Bouligand J, Guiochon-Mantel A, Brailly-Tabard S, Anderson RA, Millar RP. Kisspeptin restores pulsatile LH secretion in patients with neurokinin B signaling deficiencies: physiological, pathophysiological and therapeutic implications. Neuroendocrinology 2013; 97:193-202. [PMID: 22377698 PMCID: PMC3902960 DOI: 10.1159/000336376] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 01/05/2012] [Indexed: 11/19/2022]
Abstract
Pulsatile gonadotropin-releasing hormone (GnRH) is crucial to normal reproductive function and abnormalities in pulse frequency give rise to reproductive dysfunction. Kisspeptin and neurokinin B (NKB), neuropeptides secreted by the same neuronal population in the ventral hypothalamus, have emerged recently as critical central regulators of GnRH and thus gonadotropin secretion. Patients with mutations resulting in loss of signaling by either of these neuroendocrine peptides fail to advance through puberty but the mechanisms mediating this remain unresolved. We report here that continuous kisspeptin infusion restores gonadotropin pulsatility in patients with loss-of-function mutations in NKB (TAC3) or its receptor (TAC3R), indicating that kisspeptin on its own is sufficient to stimulate pulsatile GnRH secretion. Moreover, our findings suggest that NKB action is proximal to kisspeptin in the reproductive neuroendocrine cascade regulating GnRH secretion, and may act as an autocrine modulator of kisspeptin secretion. The ability of continuous kisspeptin infusion to induce pulsatile gonadotropin secretion further indicates that GnRH neurons are able to set up pulsatile secretion in the absence of pulsatile exogenous kisspeptin.
Collapse
Affiliation(s)
- Jacques Young
- Faculté de Médecine Paris-Sud UMR-S693, Université Paris-Sud, France
- INSERM U693, IFR93, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Service d'Endocrinologie et des Maladies de la Reproduction, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jyothis T. George
- MRC Human Reproductive Sciences Unit, University of Edinburgh, Edinburgh, UK
| | - Javier A. Tello
- MRC Human Reproductive Sciences Unit, University of Edinburgh, Edinburgh, UK
- Centre for Integrative Physiology, University of Edinburgh, School of Biomedical Sciences, Edinburgh, UK
| | - Bruno Francou
- Faculté de Médecine Paris-Sud UMR-S693, Université Paris-Sud, France
- INSERM U693, IFR93, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Jerome Bouligand
- Faculté de Médecine Paris-Sud UMR-S693, Université Paris-Sud, France
- INSERM U693, IFR93, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Anne Guiochon-Mantel
- Faculté de Médecine Paris-Sud UMR-S693, Université Paris-Sud, France
- INSERM U693, IFR93, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Sylvie Brailly-Tabard
- Faculté de Médecine Paris-Sud UMR-S693, Université Paris-Sud, France
- INSERM U693, IFR93, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Richard A. Anderson
- MRC Human Reproductive Sciences Unit, University of Edinburgh, Edinburgh, UK
| | - Robert P. Millar
- MRC Human Reproductive Sciences Unit, University of Edinburgh, Edinburgh, UK
- Centre for Integrative Physiology, University of Edinburgh, School of Biomedical Sciences, Edinburgh, UK
- Mammal Research Institute, University of Pretoria, Pretoria, and UCT/MRC Receptor Biology Unit, University of Cape Town, Cape Town, South Africa
- *Robert P. Millar, Mammal Research Institute, University of Pretoria, Pretoria 0028 (South Africa), Tel. +27 420 3776, E-Mail
| |
Collapse
|
158
|
Mahindra A, Nooney K, Uraon S, Sharma KK, Jain R. Microwave-assisted solution phase peptide synthesis in neat water. RSC Adv 2013. [DOI: 10.1039/c3ra43040e] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
159
|
Rowe ML, Elphick MR. The neuropeptide transcriptome of a model echinoderm, the sea urchin Strongylocentrotus purpuratus. Gen Comp Endocrinol 2012; 179:331-44. [PMID: 23026496 DOI: 10.1016/j.ygcen.2012.09.009] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 09/12/2012] [Accepted: 09/13/2012] [Indexed: 01/03/2023]
Abstract
Neuronal secretion of peptide signaling molecules (neuropeptides) is an evolutionarily ancient feature of nervous systems. Here we report the identification of 20 cDNAs encoding putative neuropeptide precursors in the sea urchin Strongylocentrotus purpuratus (Phylum Echinodermata), providing new insights on the evolution and diversity of neuropeptides. Identification of a gonadotropin-releasing hormone-like peptide precursor (SpGnRHP) is consistent with the widespread phylogenetic distribution of GnRH-type neuropeptides in the bilateria. A protein (SpTRHLP) comprising multiple copies of peptides that share structural similarity with thyrotropin-releasing hormone (TRH) is the first TRH-like precursor to be identified in an invertebrate. SpCTLP is the first calcitonin-like peptide with two N-terminally located cysteine residues to be found in a non-chordate species. Discovery of two proteins (SpPPLNP1, SpPPLNP2) comprising homologs of molluscan pedal peptides and arthropod orcokinins indicates the existence of a bilaterian family of pedal peptide/orcokinin-type neuropeptides. Other proteins identified contain peptides that do not share apparent sequence similarity with known neuropeptides. These include Spnp5, which comprises multiple copies of C-terminally amidated peptides that have an N-terminal Ala-Asn motif (AN peptides), and Spnp9, Spnp10 and Spnp12, which contain putative neuropeptides with a C-terminal Phe-amide, Ser-amide or Pro-amide, respectively. Several proteins (Spnp11, 14, 15, 16, 17, 18, 19 and 20) contain putative neuropeptides with multiple cysteine residues (2, 6 or 8), which may mediate formation of intramolecular or intermolecular disulphide bridges. Looking ahead, the identification of these neuropeptide precursors in S. purpuratus has provided a strong basis for a comprehensive analysis of neuropeptide function in this model echinoderm species.
Collapse
Affiliation(s)
- Matthew L Rowe
- Queen Mary University of London, School of Biological & Chemical Sciences, Mile End Road, London E1 4NS, UK
| | | |
Collapse
|
160
|
Laimou D, Katsila T, Matsoukas J, Schally A, Gkountelias K, Liapakis G, Tamvakopoulos C, Tselios T. Rationally designed cyclic analogues of luteinizing hormone-releasing hormone: enhanced enzymatic stability and biological properties. Eur J Med Chem 2012; 58:237-47. [PMID: 23127987 DOI: 10.1016/j.ejmech.2012.09.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 09/24/2012] [Accepted: 09/27/2012] [Indexed: 11/18/2022]
Abstract
This article describes the rational design, synthesis and pharmacological properties of amide-linked cyclic analogues of Luteinizing Hormone-Releasing Hormone (LHRH) with substitutions at positions 1 (Pro), 6 (D-Leu/D-Trp), 9 (Aze) and 10 (BABA/Acp). These LHRH analogues fulfil the conformational requirements that are known in the literature (bend in the 5-8 segment) to be essential for receptor recognition and activation. Although, they are characterised by an overall low binding affinity to the LHRH-I receptor, the cyclic analogues that were studied and especially the cyclo(1-10)[Pro(1), D-Leu(6), BABA(10)] LHRH, exhibit a profoundly enhanced in vitro and in vivo stability and improved pharmacokinetics in comparison with their linear counterpart and leuprolide. Upon receptor binding, cyclo(1-10)[Pro(1), D-Leu(6), BABA(10)] LHRH causes testosterone release in C57/B16 mice (in vivo efficacy) that is comparable to that of leuprolide. Testosterone release is an acutely dose dependent effect that is blocked by the LHRH-I receptor antagonist, cetrorelix. The pharmacokinetic advantages and efficacy of cyclo(1-10)[Pro(1), D-Leu(6), BABA(10)] LHRH render this analogue a promising platform for future rational drug design studies towards the development of non-peptide LHRH mimetics.
Collapse
Affiliation(s)
- Despina Laimou
- Department of Chemistry, University of Patras, GR-26500 Patras, Greece
| | | | | | | | | | | | | | | |
Collapse
|
161
|
Terasawa E, Kenealy BP. Neuroestrogen, rapid action of estradiol, and GnRH neurons. Front Neuroendocrinol 2012; 33:364-75. [PMID: 22940545 PMCID: PMC3496051 DOI: 10.1016/j.yfrne.2012.08.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/29/2012] [Accepted: 08/15/2012] [Indexed: 02/01/2023]
Abstract
Estradiol plays a pivotal role in the control of GnRH neuronal function, hence female reproduction. A series of recent studies in our laboratory indicate that rapid excitatory actions of estradiol directly modify GnRH neuronal activity in primate GnRH neurons through GPR30 and STX-sensitive receptors. Similar rapid direct actions of estradiol through estrogen receptor beta are also described in mouse GnRH neurons. In this review, we propose two novel hypotheses as a possible physiological role of estradiol in primates. First, while ovarian estradiol initiates the preovulatory GnRH surge through interneurons expressing estrogen receptor alpha, rapid direct membrane-initiated action of estradiol may play a role in sustaining GnRH surge release for many hours. Second, locally produced neuroestrogens may contribute to pulsatile GnRH release. Either way, estradiol synthesized in interneurons in the hypothalamus may play a significant role in the control of the GnRH surge and/or pulsatility of GnRH release.
Collapse
Affiliation(s)
- Ei Terasawa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, United States.
| | | |
Collapse
|
162
|
Stevenson TJ, Hahn TP, MacDougall-Shackleton SA, Ball GF. Gonadotropin-releasing hormone plasticity: a comparative perspective. Front Neuroendocrinol 2012; 33:287-300. [PMID: 23041619 PMCID: PMC3484179 DOI: 10.1016/j.yfrne.2012.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 09/04/2012] [Accepted: 09/05/2012] [Indexed: 11/16/2022]
Abstract
Gonadotropin-releasing hormone 1 (GnRH1) is a key regulator of the reproductive neuroendocrine system in vertebrates. Recent developments have suggested that GnRH1 neurons exhibit far greater plasticity at the cellular and molecular levels than previously thought. Furthermore, there is growing evidence that sub-populations of GnRH1 neurons in the preoptic area are highly responsive to specific environmental and hormonal conditions. In this paper we discuss findings that reveal large variation in GnRH1 mRNA and protein expression that are regulated by social cues, photoperiod, and hormonal feedback. We draw upon studies using histochemistry and immediate early genes (e.g., c-FOS/ZENK) to illustrate that specific groups of GnRH1 neurons are topographically organized. Based on data from diverse vertebrate species, we suggest that GnRH1 expression within individuals is temporally dynamic and this plasticity may be evolutionarily conserved. We suggest that the plasticity observed in other neuropeptide systems (i.e. kisspeptin) may have evolved in a similar manner.
Collapse
Affiliation(s)
- T J Stevenson
- Institute for Mind and Biology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
163
|
Sharma TP, Nett TM, Karsch FJ, Phillips DJ, Lee JS, Herkimer C, Padmanabhan V. Neuroendocrine control of FSH secretion: IV. Hypothalamic control of pituitary FSH-regulatory proteins and their relationship to changes in FSH synthesis and secretion. Biol Reprod 2012; 86:171. [PMID: 22423050 PMCID: PMC3386145 DOI: 10.1095/biolreprod.111.098442] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 01/04/2012] [Accepted: 02/27/2012] [Indexed: 11/01/2022] Open
Abstract
The current dogma is that the differential regulation of luteinizing hormone (LH) and follicle-stimulating hormone (FSH) synthesis and secretion is modulated by gonadotropin-releasing hormone (GnRH) pulse frequency and by changes in inhibins, activins, and follistatins both at the pituitary and at the peripheral level. To date no studies have looked at the overlapping function of these regulators in a combined setting. We tested the hypothesis that changes in GnRH pulse frequency alter the relative abundance of these regulators at the pituitary and peripheral levels in a manner consistent with changes in pituitary and circulating concentrations of FSH; that is, an increase in FSH will be accompanied by increased stimulatory input (activin) and/or reduced follistatin and inhibin. Ovariectomized ewes were subjected to a combination hypothalamic pituitary disconnection (HPD)-hypophyseal portal blood collection procedure. Hypophyseal portal and jugular blood samples were collected for a 6-h period from non-HPD ewes, HPD ewes, or HPD ewes administered GnRH hourly or every 3 h for 4 days. In the absence of endogenous hypothalamic and ovarian hormones that regulate gonadotropin secretion, 3-hourly pulses of GnRH increased pituitary content of FSH more than hourly GnRH, although these differences were not evident in the peripheral circulation. The results failed to support the hypothesis in that the preferential increase of pituitary content of FSH by the lower GnRH pulse frequency could be explained by changes in the pituitary content of inhibin A, follistatin, or activin B. Perhaps the effects of GnRH pulse frequency on FSH is due to changes in the balance of free versus bound amounts of these FSH regulatory proteins or to the involvement of other regulators not monitored in this study.
Collapse
Affiliation(s)
- Tejinder P. Sharma
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| | - Terry M. Nett
- Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, Colorado
| | - Fred J. Karsch
- Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| | - David J. Phillips
- Monash Institute of Medical Research, Monash University, Clayton, Victoria, Australia
| | - James S. Lee
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Carol Herkimer
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
- Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
164
|
Hansel W. From hyperkeratosis to apoptosis: lessons learned from 65 years of research. Biol Reprod 2012; 87:19, 1-11. [PMID: 22517625 DOI: 10.1095/biolreprod.112.100305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
|
165
|
New and emerging agents for the treatment of castration-resistant prostate cancer. Urol Oncol 2012; 29:S1-8. [PMID: 22074657 DOI: 10.1016/j.urolonc.2011.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 08/24/2011] [Accepted: 08/24/2011] [Indexed: 11/20/2022]
Abstract
Most men with recurrent prostate cancer (CaP) initially respond to androgen deprivation therapy but eventually develop metastatic castration-resistant prostate cancer (CRPC). Over the last decade, new therapeutic targets have been identified in CRPC and several new drugs have reached advanced stages of clinical development. In 2010, the Food and Drug Administration (FDA) approved sipuleucel-T and cabazitaxel, and in 2011, abiraterone for patients with metastatic CRPC based on phase 3 trials showing improved survival. Although not yet available for clinical use, a press release in June 2011 announced that radium 223 also demonstrated a survival advantage in men with metastatic CRPC. Emerging therapies in advanced stages of clinical development in CRPC include the hormonal therapies MDV3100 and TAK 700, and the immunotherapy ipilimumab. Results are also pending on phase 3 studies comparing docetaxel plus prednisone with docetaxel given with the novel agents aflibercept, dasatinib, lenalidomide, and custirsen. In addition to these new and emerging therapeutic agents, denosumab was approved for the prevention of skeletal complications in patients with bone metastases due to solid tumor malignancies, providing an alternative to zoledronic acid. While the addition of these new treatment options is a great advance for men with metastatic CRPC, there are many new questions arising regarding sequencing of these treatments with each other, with previously existing therapies, and with the emerging agents now in clinical trials. Furthermore, there are concerns that on-going phase 3 trials may be contaminated if patients go off study treatment to start 1 of the newly approved agents or take the agent subsequently. These realities make clinical trial design more challenging than ever.
Collapse
|
166
|
Servili A, Herrera-Pérez P, Kah O, Muñoz-Cueto JA. The retina is a target for GnRH-3 system in the European sea bass, Dicentrarchus labrax. Gen Comp Endocrinol 2012; 175:398-406. [PMID: 22138555 DOI: 10.1016/j.ygcen.2011.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Revised: 10/21/2011] [Accepted: 11/11/2011] [Indexed: 12/11/2022]
Abstract
The European sea bass expresses three GnRH (Gonadotrophin Releasing Hormone) forms that exert pleiotropic actions via several classes of receptors. The GnRH-1 form is responsible for the endogenous regulation of gonadotrophin release by the pituitary gland but the role of GnRH-2 and GnRH-3 remains unclear in fish. In a previous study performed in sea bass, we have provided evidence of direct links between the GnRH-2 cells and the pineal organ and demonstrated a functional role for GnRH-2 in the modulation of the secretory activity of this photoreceptive organ. In this study, we have investigated the possible relationship between the GnRH-3 system and the retina in the same species. Thus, using a biotinylated dextran-amine tract-tracing method, we reveal the presence of retinopetal cells in the terminal nerve of sea bass, a region that also contains GnRH-3-immunopositive cells. Moreover, GnRH-3-immunoreactive fibers were observed at the boundary between the inner nuclear and the inner plexiform layers, and also within the ganglion cell layer. These results strongly suggest that the GnRH-3 neurons located in the terminal nerve area represent the source of GnRH-3 innervation in the retina of this species. In order to clarify whether the retina is a target for GnRH, the expression pattern of GnRH receptors (dlGnRHR) was also analyzed by RT-PCR and in situ hybridization. RT-PCR revealed the retinal expression of dlGnRHR-II-2b, -1a, -1b and -1c, while in situ hybridization only showed positive signals for the receptors dlGnRHR-II-2b and -1a. Finally, double-immunohistochemistry showed that GnRH-3 projections reaching the sea bass retina end in close proximity to tyrosine hydroxylase (dopaminergic) cells, which also expressed the dlGnRHR-II-2b receptor subtype. Taken together, these results suggest an important role for GnRH-3 in the modulation of dopaminergic cell activities and retinal functions in sea bass.
Collapse
Affiliation(s)
- Arianna Servili
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, Campus de Excelencia Internacional del Mar (CEIMAR), E-11510 Puerto Real, Spain
| | | | | | | |
Collapse
|
167
|
Kim DK, Cho EB, Moon MJ, Park S, Hwang JI, Do Rego JL, Vaudry H, Seong JY. Molecular Coevolution of Neuropeptides Gonadotropin-Releasing Hormone and Kisspeptin with their Cognate G Protein-Coupled Receptors. Front Neurosci 2012; 6:3. [PMID: 22291614 PMCID: PMC3265131 DOI: 10.3389/fnins.2012.00003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/05/2012] [Indexed: 12/12/2022] Open
Abstract
The neuropeptides gonadotropin-releasing hormone (GnRH) and kisspeptin (KiSS), and their receptors gonadotropin-releasing hormone receptor (GnRHR) and kisspeptin receptor (KiSSR) play key roles in vertebrate reproduction. Multiple paralogous isoforms of these genes have been identified in various vertebrate species. Two rounds of genome duplication in early vertebrates likely contributed to the generation of these paralogous genes. Genome synteny and phylogenetic analyses in a variety of vertebrate species have provided insights into the evolutionary origin of and relationship between paralogous genes. The paralogous forms of these neuropeptides and their receptors have coevolved to retain high selectivity of the ligand–receptor interaction. These paralogous forms have become subfunctionalized, neofunctionalized, or dysfunctionalized during evolution. This article reviews the evolutionary mechanism of GnRH/GnRHR and KiSS/KiSSR, and the fate of the duplicated paralogs in vertebrates.
Collapse
Affiliation(s)
- Dong-Kyu Kim
- Graduate School of Medicine, Korea University Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
168
|
Schlage P, Mező G, Orbán E, Bősze S, Manea M. Anthracycline-GnRH derivative bioconjugates with different linkages: Synthesis, in vitro drug release and cytostatic effect. J Control Release 2011; 156:170-8. [DOI: 10.1016/j.jconrel.2011.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 07/30/2011] [Accepted: 08/05/2011] [Indexed: 01/13/2023]
|
169
|
Miwa K, Hitaka T, Imada T, Sasaki S, Yoshimatsu M, Kusaka M, Tanaka A, Nakata D, Furuya S, Endo S, Hamamura K, Kitazaki T. Discovery of 1-{4-[1-(2,6-difluorobenzyl)-5-[(dimethylamino)methyl]-3-(6-methoxypyridazin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-6-yl]phenyl}-3-methoxyurea (TAK-385) as a potent, orally active, non-peptide antagonist of the human gonadotropin-releasing hormone receptor. J Med Chem 2011; 54:4998-5012. [PMID: 21657270 DOI: 10.1021/jm200216q] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We previously discovered an orally active human gonadotropin-releasing hormone (GnRH) receptor antagonist, thieno[2,3-d]pyrimidine-2,4-dione derivative 1 (sufugolix). To reduce the cytochrome P450 (CYP) inhibitory activity and improve in vivo GnRH antagonistic activity, further optimization of this scaffold was carried out. We focused our synthetic efforts on chemical modification at the 5 and 3 positions of the thieno[2,3-d]pyrimidine-2,4-dione ring based on computational modeling, which resulted in the discovery of 1-{4-[1-(2,6-difluorobenzyl)-5-[(dimethylamino)methyl]-3-(6-methoxypyridazin-3-yl)-2,4-dioxo-1,2,3,4-tetrahydrothieno[2,3-d]pyrimidin-6-yl]phenyl}-3-methoxyurea (16b) as a highly potent and orally active GnRH antagonist. Compound 16b showed potent in vitro GnRH antagonistic activity in the presence of fetal bovine serum (FBS) without CYP inhibition. Oral administration of 16b maintained the suppressive effect of the plasma luteinizing hormone levels in castrated cynomolgus monkeys at a 3 mg/kg dose for more than 24 h. Compound 16b is currently under clinical development with the code name of TAK-385.
Collapse
Affiliation(s)
- Kazuhiro Miwa
- CMC Center, Takeda Pharmaceutical Company, Ltd., 17-85, Jusohonmachi 2-chome, Yodogawa-ku, Osaka 532-8686, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
170
|
Kastin AJ, Pan W. Concepts for biologically active peptides. Curr Pharm Des 2011; 16:3390-400. [PMID: 20726835 DOI: 10.2174/138161210793563491] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Accepted: 08/12/2010] [Indexed: 12/21/2022]
Abstract
Here we review a unique aspect of CNS research on biologically active peptides that started against a background of prevalent dogmas but ended by exerting considerable influence on the field. During the course of refuting some doctrines, we introduced several concepts that were unconventional and paradigm-shifting at the time. We showed that (1) hypothalamic peptides can act 'up' on the brain as well as 'down' on the pituitary, (2) peripheral peptides can affect the brain, (3) peptides can cross the blood-brain barrier, (4) the actions of peptides can persist longer than their half-lives in blood, (5) perinatal administration of peptides can exert actions persisting into adulthood, (6) a single peptide can have more than one action, (7) dose-response relationships of peptides need not be linear, (8) the brain produces antiopiate as well as opiate peptides, (9) there is a selective high affinity endogenous peptide ligand for the mu-opiate receptor, (10) a peptide's name does not restrict its effects, and (11) astrocytes assume an active role in response to metabolic disturbance and hyperleptinemia. The evolving questions in our laboratories reflect the diligent effort of the neuropeptide community to identify the roles of peptides in the CNS. The next decade is expected to see greater progress in the following areas: (a) interactions of peptides with other molecules in the CNS; (b) peptide involvement in cell-cell interactions; and (c) peptides in neuropsychiatric, autoimmune, and neurodegenerative diseases. The development of peptidomics and gene silencing approaches will expedite the formation of many new concepts in a new era.
Collapse
Affiliation(s)
- Abba J Kastin
- Blood-Brain Barrier Group, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | | |
Collapse
|
171
|
Xu Y, Faulkner LD, Hill JW. Cross-Talk between Metabolism and Reproduction: The Role of POMC and SF1 Neurons. Front Endocrinol (Lausanne) 2011; 2:98. [PMID: 22649394 PMCID: PMC3355979 DOI: 10.3389/fendo.2011.00098] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Accepted: 11/22/2011] [Indexed: 01/22/2023] Open
Abstract
Energy homeostasis and reproduction require tight coordination, but the mechanisms underlying their interaction are not fully understood. Two sets of hypothalamic neurons, namely pro-opiomelanocortin (POMC) neurons in the arcuate nucleus and steroidogenic factor-1 (SF1) neurons in the ventromedial hypothalamic nucleus, are emerging as critical nodes where metabolic and reproductive signals communicate. This view is supported by recent genetic studies showing that disruption of metabolic signals (e.g., leptin and insulin) or reproductive signals (e.g., estradiol) in these neurons leads to impaired regulation of both energy homeostasis and fertility. In this review, we will examine the potential mechanisms of neuronal communication between POMC, SF1, and gonadotropin-releasing hormone neurons in the regulation of metabolism and reproduction.
Collapse
Affiliation(s)
- Yong Xu
- Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of MedicineHouston, TX, USA
- *Correspondence: Yong Xu, Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA e-mail: ; Jennifer W. Hill, Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, College of Medicine, The University of Toledo, Toledo, OH, USA e-mail:
| | - Latrice D. Faulkner
- Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, College of Medicine, The University of ToledoToledo, OH, USA
| | - Jennifer W. Hill
- Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, College of Medicine, The University of ToledoToledo, OH, USA
- Department of Obstetrics and Gynecology, College of Medicine, The University of ToledoToledo, OH, USA
- *Correspondence: Yong Xu, Department of Pediatrics, Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX 77030, USA e-mail: ; Jennifer W. Hill, Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, College of Medicine, The University of Toledo, Toledo, OH, USA e-mail:
| |
Collapse
|
172
|
Mann RM, Hyne RV, Choung CB. Hormonal induction of spermiation, courting behavior and spawning in the southern bell frog, Litoria raniformis. Zoo Biol 2010; 29:774-82. [DOI: 10.1002/zoo.20331] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
173
|
Siejka A, Schally AV, Block NL, Barabutis N. Mechanisms of inhibition of human benign prostatic hyperplasia in vitro by the luteinizing hormone-releasing hormone antagonist cetrorelix. BJU Int 2010; 106:1382-8. [PMID: 20151966 DOI: 10.1111/j.1464-410x.2010.09215.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the mechanism by which the luteinizing hormone-releasing hormone (LHRH) antagonist cetrorelix exerts its effects in men with benign prostatic hyperplasia (BPH), as it produces a long-lasting improvement in lower urinary tract symptoms that is only partly accounted for by the transient reduction in testosterone levels, and the beneficial results could be due to direct inhibitory effects of cetrorelix on the prostate exerted through prostatic LHRH receptors. MATERIALS AND METHODS Using the BPH-1 cell line we evaluated the effects of cetrorelix in vitro on the proliferation and the expression of receptors for LHRH, epidermal growth factor (EGF), α(1A) -adrenergic receptor, STAT-3 transcription factor and the response to growth factors insulin-like growth factor (IGF)-1 and -II and fibroblast growth factor (FGF)-2. RESULTS There was expression of LHRH receptors in the human BPH-1 cell line. Cetrorelix had inhibitory effects on the proliferation rate of BPH-1 cells, also reflected by the decrease in the expression of the proliferating cell nuclear antigen (PCNA). Cetrorelix inhibited the stimulatory effect of the growth factors IGF-I and -II and FGF-2 on the proliferation of this line. Cetrorelix also downregulated the expression of the receptors for LHRH and EGF, as well as of α(1A) -adrenergic receptors, and inhibited the activation of the STAT3 transcription factor. CONCLUSIONS The results show that in vitro cetrorelix can directly inhibit the proliferation rate of the human BPH-1 cell line by counteracting growth factors like IGF-I and -II and FGF-2, and downregulating the LHRH receptor and α-adrenergic receptors, as well as transcription factors.
Collapse
Affiliation(s)
- Agnieszka Siejka
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL, USA
| | | | | | | |
Collapse
|
174
|
Abstract
The oestrous cycle in cattle lasts for 18-24 days. It consists of a luteal phase (14-18 days) and a follicular phase (4-6 days). During the cycle there are generally two (dairy cows) or three (heifers and beef cows) waves of ovarian follicle growth. Each wave of follicle growth consists of a period of emergence of a cohort of follicles, selection of a dominant follicle and either atresia or ovulation of the dominant follicle. These waves of follicle growth, initially established during the early pre-pubertal period of development occur throughout the entire cycle, with only the dominant follicle (DF) of the final wave coinciding with the follicular phase that undergoes final maturation and ovulation. Ovarian functions (follicle growth, ovulation, luteinisation and luteolysis) are regulated by the endocrine hormones of the hypothalamus (gonadotrophin-releasing hormone), anterior pituitary (follicle-stimulating hormone and luteinising hormone), ovaries (progesterone, oestradiol and inhibins) and the uterus (prostaglandin F2α). In postpartum cows resumption of regular oestrous cycles (in addition to uterine involution) is fundamental for re-establishment of pregnancy.
Collapse
|
175
|
McGuire NL, Bentley GE. Neuropeptides in the gonads: from evolution to pharmacology. Front Pharmacol 2010; 1:114. [PMID: 21607065 PMCID: PMC3095369 DOI: 10.3389/fphar.2010.00114] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Accepted: 08/07/2010] [Indexed: 01/26/2023] Open
Abstract
Vertebrate gonads are the sites of synthesis and binding of many peptides that were initially classified as neuropeptides. These gonadal neuropeptide systems are neither well understood in isolation, nor in their interactions with other neuropeptide systems. Further, our knowledge of the control of these gonadal neuropeptides by peripheral hormones that bind to the gonads, and which themselves are under regulation by true neuropeptide systems from the hypothalamus, is relatively meager. This review discusses the existence of a variety of neuropeptides and their receptors which have been discovered in vertebrate gonads, and the possible way in which such systems could have evolved. We then focus on two key neuropeptides for regulation of the hypothalamo-pituitary-gonadal axis: gonadotropin-releasing hormone (GnRH) and gonadotropin-inhibitory hormone (GnIH). Comparative studies have provided us with a degree of understanding as to how a gonadal GnRH system might have evolved, and they have been responsible for the discovery of GnIH and its gonadal counterpart. We attempt to highlight what is known about these two key gonadal neuropeptides, how their actions differ from their hypothalamic counterparts, and how we might learn from comparative studies of them and other gonadal neuropeptides in terms of pharmacology, reproductive physiology and evolutionary biology.
Collapse
Affiliation(s)
- Nicolette L McGuire
- Laboratory of Reproductive Neuroendocrinology, Department of Integrative Biology, University of California at Berkeley Berkeley, CA, USA
| | | |
Collapse
|
176
|
Khan KN, Kitajima M, Hiraki K, Fujishita A, Nakashima M, Ishimaru T, Masuzaki H. Cell proliferation effect of GnRH agonist on pathological lesions of women with endometriosis, adenomyosis and uterine myoma. Hum Reprod 2010; 25:2878-90. [DOI: 10.1093/humrep/deq240] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
177
|
Beyerman HC, Hindriks H, Hirt J, de Leer EWB, van der Wiele A. A synthesis of the decapeptide sequence proposed for the LH- and FSH-releasing hormone. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/recl.19720911011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
178
|
Lehman MN, Coolen LM, Goodman RL. Minireview: kisspeptin/neurokinin B/dynorphin (KNDy) cells of the arcuate nucleus: a central node in the control of gonadotropin-releasing hormone secretion. Endocrinology 2010; 151:3479-89. [PMID: 20501670 PMCID: PMC2940527 DOI: 10.1210/en.2010-0022] [Citation(s) in RCA: 551] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, a subset of neurons was identified in the arcuate nucleus of the hypothalamus that colocalize three neuropeptides, kisspeptin, neurokinin B, and dynorphin, each of which has been shown to play a critical role in the central control of reproduction. Growing evidence suggests that these neurons, abbreviated as the KNDy subpopulation, are strongly conserved across a range of species from rodents to humans and play a key role in the physiological regulation of GnRH neurons. KNDy cells are a major target for steroid hormones, form a reciprocally interconnected network, and have direct projections to GnRH cell bodies and terminals, features that position them well to convey steroid feedback control to GnRH neurons and potentially serve as a component of the GnRH pulse generator. In addition, recent work suggests that alterations in KNDy cell peptides may underlie neuroendocrine defects seen in clinical reproductive disorders such as polycystic ovarian syndrome. Taken together, this evidence suggests a key role for the KNDy subpopulation as a focal point in the control of reproductive function in health and disease.
Collapse
Affiliation(s)
- Michael N Lehman
- Department of Anatomy and Cell Biology, Medical Sciences Building, University of Western Ontario, London, Ontario, Canada.
| | | | | |
Collapse
|
179
|
Terasawa E, Kurian JR, Guerriero KA, Kenealy BP, Hutz ED, Keen KL. Recent discoveries on the control of gonadotrophin-releasing hormone neurones in nonhuman primates. J Neuroendocrinol 2010; 22:630-8. [PMID: 20456608 PMCID: PMC2908205 DOI: 10.1111/j.1365-2826.2010.02027.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Since Ernst Knobil proposed the concept of the gonadotrophin-releasing hormone (GnRH) pulse-generator in the monkey hypothalamus three decades ago, we have made significant progress in this research area with cellular and molecular approaches. First, an increase in pulsatile GnRH release triggers the onset of puberty. However, the question of what triggers the pubertal increase in GnRH is still unclear. GnRH neurones are already mature before puberty but GnRH release is suppressed by a tonic GABA inhibition. Our recent work indicates that blocking endogenous GABA inhibition with the GABA(A) receptor blocker, bicuculline, dramatically increases kisspeptin release, which plays an important role in the pubertal increase in GnRH release. Thus, an interplay between the GABA, kisspeptin, and GnRH neuronal systems appears to trigger puberty. Second, cultured GnRH neurones derived from the olfactory placode of monkey embryos exhibit synchronised intracellular calcium, [Ca(2+)](i), oscillations and release GnRH in pulses at approximately 60-min intervals after 14 days in vitro (div). During the first 14 div, GnRH neurones undergo maturational changes from no [Ca(2+)](i) oscillations and little GnRH release to the fully functional state. Recent work also shows GnRH mRNA expression increases during in vitro maturation. This mRNA increase coincides with significant demethylation of a CpG island in the GnRH 5'-promoter region. This suggests that epigenetic differentiation occurs during GnRH neuronal maturation. Third, oestradiol causes rapid, direct, excitatory action in GnRH neurones and this action of oestradiol appears to be mediated through a membrane receptor, such as G-protein coupled receptor 30.
Collapse
Affiliation(s)
- E Terasawa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715-1299, USA.
| | | | | | | | | | | |
Collapse
|
180
|
Vera MD, Lundquist JT, Chengalvala MV, Cottom JE, Feingold IB, Garrick LM, Green DM, Hauze DB, Mann CW, Mehlmann JF, Rogers JF, Shanno L, Wrobel JE, Pelletier JC. Synthesis and biological evaluation of piperazinyl heterocyclic antagonists of the gonadotropin releasing hormone (GnRH) receptor. Bioorg Med Chem Lett 2010; 20:2512-5. [DOI: 10.1016/j.bmcl.2010.02.099] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2010] [Accepted: 02/26/2010] [Indexed: 10/19/2022]
|
181
|
|
182
|
Abstract
BACKGROUND Studies suggest that testosterone (TT) replacement may have an antidepressant effect in depressed patients. OBJECTIVE The objective of this study was to explore the effect of TT administration on depression using both a systematic review of the literature and a meta-analysis. METHODOLOGY A search was conducted of MEDLINE, the Clinical Trials Registry, and Cochrane Central for English-language publications concerning randomized, placebo-controlled trials involving use of TT therapy in depressed patients. We searched for additional trials in the individual reference lists of the articles identified in the search. A study was judged to be relevant for inclusion in this review and meta-analysis if it reported original data from a controlled trial comparing use of TT and placebo in patients diagnosed with a depressive disorder according to DSM criteria, and the treatment response was evaluated according to changes on the Hamilton Rating Scale for Depression (HAM-D). We extracted the following data from the identified studies: study source, total number of participants in the study and in each treatment group, participants' ages, number of participants with a diagnosis of hypogonadism or HIV/AIDS, study duration, type of intervention, and change in HAM-D scores in the groups receiving TT versus placebo. The meta-analysis evaluated the effect of TT replacement on response in depressed patients as measured by change in HAM-D scores in the available placebo-controlled, randomized clinical trails. RESULTS Seven studies (N=364) were identified that included a placebo-control group in a double-blind design. Eligibility criteria were clearly reported in all trials. Meta-analysis of the data from these seven studies showed a significant positive effect of TT therapy on HAM-D response in depressed patients when compared with placebo (z=4.04, P<0.0001). Subgroup analysis also showed a significant response in the subpopulations with hypogonadism (z=3.84, P=0.0001) and HIV/AIDS (z=3.33, P=0.0009) as well as in patients treated with TT gel (z=2.32, P=0.02). CONCLUSIONS TT may have an antidepressant effect in depressed patients, especially those with hypogonadism or HIV/AIDS and elderly subpopulations. The route by which TT is administered may play a role in treatment response.
Collapse
|
183
|
Szabó I, Manea M, Orbán E, Csámpai A, Bosze S, Szabó R, Tejeda M, Gaál D, Kapuvári B, Przybylski M, Hudecz F, Mezo G. Development of an oxime bond containing daunorubicin-gonadotropin-releasing hormone-III conjugate as a potential anticancer drug. Bioconjug Chem 2009; 20:656-65. [PMID: 19296605 DOI: 10.1021/bc800542u] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Here, we report on the synthesis and biological properties of a conjugate in which daunorubicin (Dau) as chemotherapeutic agent was attached through an oxime bond to gonadotropin-releasing hormone-III (GnRH-III) as targeting moiety. In vitro toxicity and the cytostatic effect of the conjugate on MCF-7 human breast and C26 murine colon cancer cell lines were determined, and the results were compared with those obtained for the free daunorubicin, as well as with the doxorubicin containing derivative. In vivo antitumor effect of daunorubicin-GnRH-III was studied on Balb/c female mice transplanted with C26 tumor. Our data indicate that the daunorubicin-GnRH-III conjugate had a lower toxic effect than the free daunorubicin and it was essentially nontoxic up to 15 mg (Dau content)/kg body weight. The treatment of the C26 tumor bearing mice with the conjugate led to tumor growth inhibition and longer survival time in comparison with the controls and with the administration of the free drug. When mice were treated twice with the conjugate (on days 4 and 7 after tumor transplantation), 46% tumor growth inhibition was obtained. In this case, the increase of the median survival time was 38% compared to the controls.
Collapse
Affiliation(s)
- Ildikó Szabó
- Hungarian Academy of Sciences, and Institute of Chemistry, Eotvos Lorand University, Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Magee C, Foradori CD, Bruemmer JE, Arreguin-Arevalo JA, McCue PM, Handa RJ, Squires EL, Clay CM. Biological and anatomical evidence for kisspeptin regulation of the hypothalamic-pituitary-gonadal axis of estrous horse mares. Endocrinology 2009; 150:2813-21. [PMID: 19228887 DOI: 10.1210/en.2008-1698] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The purpose of the present study was to evaluate the effects of kisspeptin (KiSS) on LH and FSH secretion in the seasonally estrous mare and to examine the distribution and connectivity of GnRH and KiSS neurons in the equine preoptic area (POA) and hypothalamus. The diestrous mare has a threshold serum gonadotropin response to iv rodent KiSS decapeptide (rKP-10) administration between 1.0 and 500 microg. Administration of 500 microg and 1.0 mg rKP-10 elicited peak, mean, and area under the curve LH and FSH responses indistinguishable to that of 25 microg GnRH iv, although a single iv injection of 1.0 mg rKP-10 was insufficient to induce ovulation in the estrous mare. GnRH and KiSS-immunoreactive (ir) cells were identified in the POA and hypothalamus of the diestrous mare. In addition, KiSS-ir fibers were identified in close association with 33.7% of GnRH-ir soma, suggesting a direct action of KiSS on GnRH neurons in the mare. In conclusion, we are the first to reveal a physiological role for KiSS in the diestrous mare with direct anatomic evidence by demonstrating a threshold-like gonadotropin response to KiSS administration and characterizing KiSS and GnRH-ir in the POA and hypothalamus of the diestrous horse mare.
Collapse
Affiliation(s)
- Christianne Magee
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | | | | | | | | | |
Collapse
|
185
|
Abstract
SUMMARY Many hormones are secreted in pulses. The pulsatile relationship between hormones regulates many biological processes. To understand endocrine system regulation, time series of hormone concentrations are collected. The goal is to characterize pulsatile patterns and associations between hormones. Currently each hormone on each subject is fitted univariately. This leads to estimates of the number of pulses and estimates of the amount of hormone secreted; however, when the signal-to-noise ratio is small, pulse detection and parameter estimation remains difficult with existing approaches. In this article, we present a bivariate deconvolution model of pulsatile hormone data focusing on incorporating pulsatile associations. Through simulation, we exhibit that using the underlying pulsatile association between two hormones improves the estimation of the number of pulses and the other parameters defining each hormone. We develop the one-to-one, driver-response case and show how birth-death Markov chain Monte Carlo can be used for estimation. We exhibit these features through a simulation study and apply the method to luteinizing and follicle stimulating hormones.
Collapse
Affiliation(s)
- Nichole E Carlson
- Department of Biostatistics and Informatics, University of Colorado-Denver, Denver, Colorado 80262, USA.
| | | | | |
Collapse
|
186
|
Affiliation(s)
- Judd W Moul
- Division of Urologic Surgery, Department of Surgery, Duke Prostate Center, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
187
|
Pelletier JC, Chengalvala MV, Cottom JE, Feingold IB, Green DM, Hauze DB, Huselton CA, Jetter JW, Kopf GS, Lundquist JT, Magolda RL, Mann CW, Mehlmann JF, Rogers JF, Shanno LK, Adams WR, Tio CO, Wrobel JE. Discovery of 6-({4-[2-(4-tert-Butylphenyl)-1H-benzimidazol-4-yl]piperazin-1-yl}methyl)quinoxaline (WAY-207024): An Orally Active Antagonist of the Gonadotropin Releasing Hormone Receptor (GnRH-R). J Med Chem 2009; 52:2148-52. [DOI: 10.1021/jm801572m] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jeffrey C. Pelletier
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Murty V. Chengalvala
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Joshua E. Cottom
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Irene B. Feingold
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Daniel M. Green
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Diane B. Hauze
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Christine A. Huselton
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - James W. Jetter
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Gregory S. Kopf
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Joseph T. Lundquist
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Ronald L. Magolda
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Charles W. Mann
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - John F. Mehlmann
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - John F. Rogers
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Linda K. Shanno
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - William R. Adams
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Cesario O. Tio
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Jay E. Wrobel
- Department of Chemical Sciences, Musculoskeletal Biology, and Drug Safety and Metabolism, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| |
Collapse
|
188
|
Nair RM, Schally AV. Structure of a hypothalamic peptide possessing gonadotropin-releasing activity. INTERNATIONAL JOURNAL OF PEPTIDE AND PROTEIN RESEARCH 2009; 4:421-30. [PMID: 4569573 DOI: 10.1111/j.1399-3011.1972.tb03449.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
189
|
Abstract
Androgen privation is considered as the referent first line treatment for metastatic prostate cancer. Based on LHRH agonist, different therapeutic schedule included maximum androgenic blockage, intermittent treatment and associations with other drugs like oestrogen leading to possible hormonal manipulations. Since metastasis is confirmed, immediate treatment with continue LHRH agonist is the French Association of Urology (AFU) AFU recommendations treatment for metastatic prostate cancer but intermittent treatment can be considered as an option.
Collapse
|
190
|
Green DM, Goljer I, Andraka DS, Chengalvala M, Shanno L, Hurlburt W, Pelletier JC. Parallel Synthesis of 2-Aryl-4-aminobenzimidazoles and Their Evaluation as Gonadotropin Releasing Hormone Antagonists. ACTA ACUST UNITED AC 2008; 11:117-25. [DOI: 10.1021/cc800106h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Daniel M. Green
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Igor Goljer
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Diane S. Andraka
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Murty Chengalvala
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Linda Shanno
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Warren Hurlburt
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| | - Jeffrey C. Pelletier
- Departments of Chemical And Screeing Sciences and Women’s Health & Musculoskeletal Biology, Wyeth Research, 500 Arcola Road, Collegeville, Pennsylvania 19426
| |
Collapse
|
191
|
Checura CM, Beg MA, Gastal EL, Gastal MO, Wiltbank MC, Parrish JJ, Ginther OJ. Effect of suppression of FSH with a GnRH antagonist (acyline) before and during follicle deviation in the mare. Reprod Domest Anim 2008; 44:504-11. [PMID: 18954386 DOI: 10.1111/j.1439-0531.2008.01222.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A GnRH antagonist (Acyline) was used to study the role of FSH in early development of a follicular wave in 61 mares. In Experiment 1, a single dose of 3 mg per mare, compared with 0 and 1 mg, suppressed both the FSH and follicle responses to exogenous GnRH. In Experiment 2, high concentrations of FSH were induced by two successive ablations of all follicles >/= 6 mm on days 10 and 13 (day 0 = ovulation). A single treatment with Acyline resulted in significantly greater suppression of plasma concentrations of FSH than a single treatment with charcoal-extracted follicular fluid (source of inhibin) or oestradiol. Suppression of FSH was not significantly different between the group treated with Acyline alone and a group treated with a combination of Acyline, inhibin and oestradiol. In Experiment 3, all follicles were ablated on day 10 to induce an FSH surge and a new follicular wave. Acyline treatment on day 10 resulted in an immediate decrease in FSH, without a significant effect on day of emergence of a new wave or growth of follicles from 7 to 11 mm on days 11-13. Treatment on day 15, a day before expected follicle deviation and after the peak of the wave-stimulating FSH surge, resulted in an immediate decrease in FSH and cessation of follicle growth. Results indicated that growth of follicles for about 2 days after wave emergence was independent of FSH. In contrast, during the decline in the wave-stimulating FSH surge and before follicle deviation, growth of follicles was dependent on FSH.
Collapse
Affiliation(s)
- C M Checura
- Eutheria Foundation, Cross Plains, WI 53528, USA
| | | | | | | | | | | | | |
Collapse
|
192
|
Abstract
Breast cancer remains the second leading cause of malignancy-related death in women in the USA, regardless of advances in novel therapeutic agents. High priority should be given to research aimed at the study of pharmacological and natural compounds that could potentially prevent the development of breast cancer in susceptible patients. Tamoxifen has been shown to reduce the incidence of estrogen receptor-positive invasive breast cancer in women with a high risk of developing this condition by nearly 50%, and studies in osteoporosis have revealed a similar protective effect of raloxifene in postmenopausal women. The aromatase inhibitors are superior to tamoxifen in reducing the recurrence of breast cancer in postmenopausal women; large clinical trials are currently evaluating the chemopreventive effect of these agents. The list of agents with the potential for chemoprevention in breast cancer is extensive and continues to expand. There is an immense need to develop drugs that will decrease the incidence of estrogen receptor-negative breast cancer in women at high risk of developing the disease. Herein, we review the most important chemopreventive agents in breast cancer and clinical trials that have evaluated their efficacy.
Collapse
Affiliation(s)
- Aurelio B Castrellon
- University of Miami Miller's School Of Medicine, Division of Hematology/Medical Oncology, Sylvester Comprehensive Cancer Center, 1475 NW 12th Avenue Miami, FL 33136, USA.
| | | |
Collapse
|
193
|
Heitman LH, Ye K, Oosterom J, Ijzerman AP. Amiloride derivatives and a nonpeptidic antagonist bind at two distinct allosteric sites in the human gonadotropin-releasing hormone receptor. Mol Pharmacol 2008; 73:1808-15. [PMID: 18344315 DOI: 10.1124/mol.107.043521] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
The interest in the allosteric modulation of G protein-coupled receptors has grown during the past decade. It has been shown that ligands acting at allosteric sites present in these important drug targets have the ability to modulate receptor conformations and fine-tune pharmacological responses to the orthosteric ligand. In the present study, allosteric modulation of the human gonadotropin-releasing hormone (GnRH) receptor by amiloride analogs [e.g., 5-(N,N-hexamethylene)amiloride (HMA)] and a nonpeptide antagonistic furan derivative (FD-1) was studied. First, the compounds' ability to influence the dissociation of a radiolabeled peptide agonist ((125)I-triptorelin) from human GnRH receptors stably expressed in Chinese hamster ovary cell membranes was investigated. HMA and FD-1, but not 5-(N-benzyl-N-methylaminomethyl)1-(2,6-difluorobenzyl)-6-[4-(3-methoxyureido)phenyl]-3-phenylthieno[2,3-d]pyrimidine-2,4(1H,3H)-dione (TAK-013), another nonpeptide antagonist, were shown to increase the dissociation rate of (125)I-triptorelin, revealing their allosteric inhibitory characteristics. The simultaneous addition of HMA and FD-1 resulted in an additive effect on the dissociation rate. Second, in a functional assay, it was shown that HMA was a noncompetitive antagonist and that FD-1 had both competitive and noncompetitive antagonistic properties. Equilibrium displacement studies showed that the inhibition of (125)I-triptorelin binding by FD-1 was not affected by HMA. Furthermore, the potency of HMA to increase radioligand dissociation was not affected by the presence of FD-1. Simulation of the data obtained in the latter experiment also indicated neutral cooperativity between the binding of HMA and FD-1. Taken together, these results demonstrate that HMA and FD-1 are allosteric inhibitors that bind at two distinct, noncooperative, allosteric sites. This presence of a second allosteric site may provide yet another opportunity for the discovery of new ligands for the human GnRH receptor.
Collapse
Affiliation(s)
- Laura H Heitman
- Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, University of Leiden, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | |
Collapse
|
194
|
Tello JA, Wu S, Rivier JE, Sherwood NM. Four functional GnRH receptors in zebrafish: analysis of structure, signaling, synteny and phylogeny. Integr Comp Biol 2008; 48:570-87. [DOI: 10.1093/icb/icn070] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
195
|
Bentley GE, Ubuka T, McGuire NL, Chowdhury VS, Morita Y, Yano T, Hasunuma I, Binns M, Wingfield JC, Tsutsui K. Gonadotropin-inhibitory hormone and its receptor in the avian reproductive system. Gen Comp Endocrinol 2008; 156:34-43. [PMID: 18031743 DOI: 10.1016/j.ygcen.2007.10.003] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2007] [Revised: 10/08/2007] [Accepted: 10/10/2007] [Indexed: 11/19/2022]
Abstract
Many hormones that are classified as neuropeptides are synthesized in vertebrate gonads in addition to the brain. Receptors for these hormones are also expressed in gonadal tissue; thus there is potential for a highly localized autocrine or paracrine effect of these hormones on a variety of gonadal functions. In the present study we focused on gonadotropin-inhibitory hormone (GnIH), a neuropeptide that was first discovered in the hypothalamus of birds. We present different lines of evidence for the synthesis of GnIH and its receptor in the avian reproductive system including gonads and accessory reproductive organs by studies on two orders of birds: Passeriformes and Galliformes. Binding sites for GnIH were initially identified via in vivo and in vitro receptor fluorography, and were localized in ovarian granulosa cells along with the interstitial layer and seminiferous tubules of the testis. Furthermore, species-specific primers produced clear PCR products of GnIH and GnIH receptor (GnIH-R) in songbird and quail gonadal and other reproductive tissues, such as oviduct, epididymis and vas deferens. Sequencing of the PCR products confirmed their identities. Immunocytochemistry detected GnIH peptide in ovarian thecal and granulosa cells, testicular interstitial cells and germ cells and pseudostratified columnar epithelial cells in the epididymis. In situ hybridization of GnIH-R mRNA in testes produced a strong reaction product which was localized to the germ cells and interstitium. In the epididymis, the product was also localized in the pseudostratified columnar epithelial cells. In sum, these results indicate that the avian reproductive system has the capability to synthesize and bind GnIH in several tissues. The distribution of GnIH and its receptor suggest a potential for autocrine/paracrine regulation of gonadal steroid production and germ cell differentiation and maturation.
Collapse
Affiliation(s)
- George E Bentley
- Laboratory of Reproductive Neuroendocrinology, Department of Integrative Biology and Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, CA 94720-3140, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Coyral-Castel S, Tosca L, Ferreira G, Jeanpierre E, Rame C, Lomet D, Caraty A, Monget P, Chabrolle C, Dupont J. The effect of AMP-activated kinase activation on gonadotrophin-releasing hormone secretion in GT1-7 cells and its potential role in hypothalamic regulation of the oestrous cyclicity in rats. J Neuroendocrinol 2008; 20:335-46. [PMID: 18194429 DOI: 10.1111/j.1365-2826.2007.01643.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Hypothalamic AMP-activated kinase (AMPK) is a key regulator of food intake in mammals. Its role in reproduction at the central level and, more precisely, in gonadotrophin-releasing hormone (GnRH) release has never been investigated. We showed that each subunit of AMPK is present in immortalised GnRH neurones (GT1-7 cells). Treatment with 5-aminoimidazole-4-carboxamide-1-beta-D-ribonucleoside (AICAR) and metformin, two activators of AMPK, increased dose-dependent and time-dependent phosphorylation of AMPKalpha atThr172 in GT1-7 cells. Phosphorylation of acetyl-coenzyme A carboxylase at ser79 also increased. Treatment with AICAR (5 mM) or metformin (5 mM) for 4 h inhibited GnRH release in the presence or absence of GnRH (10(-8) M). Specific AMPK inhibitor compound C completely eliminated the effects of AICAR or metformin on GnRH release. Finally, we determined the central effects of AICAR in vivo on food intake and oestrous cyclicity. Ten-week-old female rats received a 50 microg AICAR or a saline i.c.v. injection. We detected increased AMPK and acetyl-CoA carboxylase phosphorylation, specifically in the hypothalamus, 30 min after AICAR injection. Food intake was significantly higher (P < 0.05) in animals treated with AICAR than in animals injected with saline, 24 h after injection. This effect was abolished after 1 week. Moreover, during the 4 weeks following injection, the interval between two oestrous stages was significantly lower in the AICAR group than in the saline group. Our findings suggest that AMPK activation may act directly at the hypothalamic level to affect fertility by modulating GnRH release and oestrous cyclicity.
Collapse
Affiliation(s)
- S Coyral-Castel
- Unité de Physiologie de la Reproduction et des Comportements, UMR6175, INRA, Nouzilly, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Chammas NK, Chambers SM, Harris PE. The GnRH test in the assessment of patients with pituitary and parapituitary lesions: results of a 5-year retrospective study. Pituitary 2008; 11:271-8. [PMID: 18425583 DOI: 10.1007/s11102-008-0124-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To examine the utility of the GnRH (gonadotrophin-releasing hormone) test in the management of patients with pituitary and parapituitary lesions. PATIENTS AND METHODS A 5-year retrospective study of LH (luteinizing hormone) and FSH (follicle stimulating hormone) responses to GnRH test in patients with HP (hypothalamic-pituitary) disease in a regional endocrine centre. Serum LH and FSH concentrations were measured at baseline and at 20 and 60 min after an intravenous bolus of 100 mcg (micrograms) of GnRH. The GnRH responses were categorised by tumour size, tumour type, and gonadal status. RESULTS Of the 104 patients studied, 46 were male and 58 were female. There were 50 normal, 38 subnormal and 16 exaggerated LH responses compared with 34 normal 67 subnormal and three exaggerated responses for FSH. Seventy-four patients (71.2%) were hypogonadal. Normal LH responses were achieved in half of the hypogonadal subjects and normal FSH responses in more than a third. Furthermore, the LH responses were exaggerated in nine hypogonadal patients compared with three for FSH. The GnRH test could not differentiate between pituitary or parapituitary lesions either by size or type of lesion. An exception was the male non-functioning adenoma (NFA) sub-group (10 patients, all were hypopituitary, seven were hypogonadal), which demonstrated significant subnormal LH and FSH responses compared with other male and female tumour type sub-groups. CONCLUSIONS The data from this study indicate that the GnRH test is unhelpful in the clinical assessment of the HP axis in patients with HP disease.
Collapse
Affiliation(s)
- N K Chammas
- Endocrine Unit, King's College Hospital, London, SE5 9PJ, UK.
| | | | | |
Collapse
|
198
|
Wilson AC, Vadakkadath Meethal S, Bowen RL, Atwood CS. Leuprolide acetate: a drug of diverse clinical applications. Expert Opin Investig Drugs 2007; 16:1851-63. [DOI: 10.1517/13543784.16.11.1851] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
199
|
Zerbib M. [Contribution of LHRH analogs in prostate cancer treatment]. ANNALES D'UROLOGIE 2007; 41 Suppl 3:S83-S86. [PMID: 18297907 DOI: 10.1016/s0003-4401(07)80516-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The contribution of LHRH analogs is enormous in prostate cancer management at nearly all stages of the disease, the Leading cancer in males over 50 years of age. They make it possible to use hormone therapy reliably, with Little morbidity, and can be reversed. Their use can be immediate or delayed, continuous or intermittent, whether or not they are associated with an antiandrogen, and can be associated with chemotherapy for patients who have reached the stage of hormone-refractory prostate cancer.
Collapse
Affiliation(s)
- M Zerbib
- Service d'urologie, hôpital Cochin, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France.
| |
Collapse
|
200
|
Abstract
Because gonadotropin-releasing hormone (GnRH) analogs constitute an important class of therapeutics for various reproductive and hormone-dependent disorders, many novel compounds have been discovered and studied. Several orally active nonpeptide GnRH antagonists have recently gained increased attention. In the study published in this issue of Molecular Pharmacology, Kohout et al. (p. 238) used small-molecule TAK-013 (sufugolix; developed previously by Takeda Chemical Industries) as a tool to elucidate the mechanism of its insurmountable antagonism. On the basis of receptor mutagenesis combined with molecular modeling, the authors hypothesized that certain amino acid sequences uniquely present in the human GnRH receptor amino terminus and extracellular loop 2 may form a "trap door" retarding dissociation of TAK-013. Such a trapping mechanism could be both ligand- and receptor species-specific. Although analogous models were previously proposed for other G protein-coupled receptors, the study by Kohout et al. (2007) provides an important advance in the GnRH antagonists field and an illustration of the fact that preclinical studies using animal models with nonhuman receptors may have very limited value in predicting drug efficacy in human disease. There are many examples showing that high-affinity protein, peptide, or nonpeptide agonists or antagonists have also enhanced clinical efficacy. However, there are also numerous studies indicating that very high receptor binding affinity is not a guarantee of drug efficacy and that other factors, including pharmacokinetic profile, ligand-induced receptor desensitization, and "trafficking," are critical in design and development of optimal drugs.
Collapse
|