101
|
Ortega I, García-Velasco JA, Pellicer A. Ovarian manipulation in ART: going beyond physiological standards to provide best clinical outcomes. J Assist Reprod Genet 2018; 35:1751-1762. [PMID: 30056596 DOI: 10.1007/s10815-018-1258-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022] Open
Abstract
Current knowledge on ovarian physiology has challenged the traditional concept of folliculogenesis, creating the basis for novel ovarian stimulation protocols in assisted reproduction technology. The purpose of this review was to evaluate the efficacy of novel clinical interventions that could aid clinicians in individualizing their protocols to patients' characteristics and personal situations. We conducted a literature review of the available evidence on new approaches for ovarian stimulation from both retrospective and prospective studies in the PubMed database. Here, we present some of the most important interventions, including follicle growth in the gonadotropin-independent and dependent stage, manipulation of estradiol production throughout ovarian stimulation, control of mid-cycle gonadotropin surges, and luteal phase support after different stimulation protocols and trigger agents. The latest research on IVF has moved physicians away from the classical physiology, allowing the development of new strategies to decouple organ functions from the female reproductive system and challenging the traditional concept of IVF.
Collapse
Affiliation(s)
- Israel Ortega
- IVI-Madrid, Madrid, Spain. .,Instituto de Investigación Sanitaria La Fé, Valencia, Spain.
| | - Juan A García-Velasco
- IVI-Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria La Fé, Valencia, Spain.,Rey Juan Carlos University, Madrid, Spain.,IdiPAZ, Madrid, Spain
| | - Antonio Pellicer
- Instituto de Investigación Sanitaria La Fé, Valencia, Spain.,Rey Juan Carlos University, Madrid, Spain.,IdiPAZ, Madrid, Spain.,IVI-Roma, Rome, Italy
| |
Collapse
|
102
|
Hara I, Yamashita S, Nishizawa S, Kikkawa K, Shimokawa T, Kohjimoto Y. Enzalutamide Versus Abiraterone as a First-Line Endocrine Therapy for Castration-Resistant Prostate Cancer: Protocol for a Multicenter Randomized Phase 3 Trial. JMIR Res Protoc 2018; 7:e11191. [PMID: 30054264 PMCID: PMC6086929 DOI: 10.2196/11191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/09/2018] [Indexed: 12/15/2022] Open
Abstract
Background Recent large-scale randomized studies have demonstrated that 2 new hormone preparations (abiraterone and enzalutamide) prolong survival in docetaxel-treated or -naïve castration-resistant prostate cancer patients. However, no studies have directly compared antitumor effects between these 2 agents, and no clear guidelines are available for choosing between them. Objective The objective of this clinical study is to compare antitumor effects and adverse events between abiraterone and enzalutamide by allocating castration-resistant prostate cancer patients deemed not indicated for docetaxel treatment to receive either of the 2 agents. Methods This study is an open-label, comparative study allocating castration-resistant prostate cancer patients to abiraterone or enzalutamide treatment arms (allocation factors: age <70 vs ≥70 years, and presence vs absence of metastases) and assessing the treatment results. Each arm will contain 25 patients. On confirmation of prostate-specific antigen failure or progression on imaging, patients undergo crossover to receive the alternative study drug. The primary end point is prostate-specific antigen response rate (percentage of patients with a decrease in prostate-specific antigen level by ≥50%) in the abiraterone and enzalutamide treatment arms. Results Recruitment started in May 2016, and 13 patients have been recruited so far. We expect to complete enrollment by December 2020. Conclusions Recently, cross-resistance between abiraterone and enzalutamide has been an issue of focus. Urologists thus tend to prefer docetaxel rather than sequential therapies using 2 hormonal preparations after the progression of a first hormonal preparation. From that perspective, our clinical trial is rather out of fashion. Nevertheless, we assume that many patients receive hormonal sequential therapy in the actual clinical setting, since most such patients cannot receive chemotherapeutic agents due to old age or poor performance status. This is why we are attempting this randomized clinical trial comparing abiraterone versus enzalutamide. We will try to identify which drug is suitable for initial hormonal therapy among castration-resistant prostate cancer patients who do not meet the indications for docetaxel therapy in terms of not only antitumor effect, but also adverse events and quality of life. Trial Registration University Hospital Medical Information Network UMIN000022102; https://upload.umin.ac.jp /cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000025463 (Archived by WebCite at http://www.webcitation.org/70xaQfGlJ)
Collapse
Affiliation(s)
- Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | | | | | - Kazuro Kikkawa
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Toshio Shimokawa
- Clinical Research Center, Wakayama Medical University, Wakayama, Japan
| | - Yasuo Kohjimoto
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
103
|
Haziak K, Herman AP, Wojtulewicz K, Pawlina B, Paczesna K, Bochenek J, Tomaszewska-Zaremba D. Effect of CD14/TLR4 antagonist on GnRH/LH secretion in ewe during central inflammation induced by intracerebroventricular administration of LPS. J Anim Sci Biotechnol 2018; 9:52. [PMID: 30026944 PMCID: PMC6047126 DOI: 10.1186/s40104-018-0267-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 05/17/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Immune stress induced by lipopolysaccharide (LPS) influences the gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) secretion. Presence of LPS interacting Toll-like receptor (TLR) 4 in the hypothalamus may enable the direct action of LPS on the GnRH/LH secretion. So, the aim of the study was to investigate the influence of intracerebroventricular (icv) injection of TLR4 antagonist on GnRH/LH secretion in anestrous ewes during LPS-induced central inflammation. Animals were divided into three groups icv-treated with: Ringer-Locke solution, LPS and TLR4 antagonist followed by LPS. RESULTS It was demonstrated that TLR4 antagonist reduced LPS-dependent suppression of GnRH gene expression in the preoptic area and in the medial basal hypothalamus, and suppression of receptor for GnRH gene expression in the anterior pituitary gland. It was also shown that TLR4 antagonist reduced suppression of LH release caused by icv injection of LPS. Central administration of LPS stimulated TLR4 gene expression in the medial basal hypothalamus. CONCLUSIONS It was indicated that blockade of TLR4 prevents the inhibitory effect of centrally acting LPS on the GnRH/LH secretion. This suggests that some negative effects of bacterial infection on the hypothalamic-pituitary-gonadal axis activity at the hypothalamic level may be caused by central action of LPS acting through TLR4.
Collapse
Affiliation(s)
- Karolina Haziak
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Andrzej Przemysław Herman
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Karolina Wojtulewicz
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Bartosz Pawlina
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Kamila Paczesna
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Joanna Bochenek
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| | - Dorota Tomaszewska-Zaremba
- The Kielanowski Institute of Animal Physiology and Nutrition, Polish Academy of Sciences, 05-110 Jabłonna, Poland
| |
Collapse
|
104
|
Carrasco RA, Singh J, Adams GP. Distribution and morphology of gonadotropin-releasing hormone neurons in the hypothalamus of an induced ovulator - The llama (Lama glama). Gen Comp Endocrinol 2018; 263:43-50. [PMID: 29656045 DOI: 10.1016/j.ygcen.2018.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/23/2018] [Accepted: 04/11/2018] [Indexed: 11/30/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a decapeptide involved in the regulation of reproduction in all mammals, but the distribution of GnRH neurons within the brain varies widely among species. The objective of the present study was to characterize the number and distribution of GnRH neurons in the hypothalamus and preoptic area of llamas, an induced ovulator. The brains of female llamas (n = 4) were fixed, frozen and sectioned serially every 50 µm in the transverse (coronal) plane. Every 10th section was stained for immunohistochemical detection of GnRH-positive neuron cell bodies and fibers by incubation with 3,3'-diaminobenzidine. The number of counted immunoreactive cells ranged from 222 to 250 (≈241 ± 13 cells in the preoptic area and hypothalamus per animal) and were localized in the medio-basal hypothalamus (44.3%), anterior hypothalamus (27%), preoptic area (14.9%), diagonal band of Broca/medial septum (13.4%), and mammillary area (0.5%). The immunoreactive cells were not localized in specific hypothalamic nuclei, but rather appeared to be distributed diffusely. The highest concentration of immunoreactive neuron fibers was in the median eminence (P < 0.05), but fibers were identified in most of the areas analyzed, including the neurohypophysis. The GnRH neurons within the hypothalamus displayed monopolar (33%), bipolar (39%), and multipolar (28%) morphologies. The bipolar type was most common in the medio-basal region (40%; P < 0.05). We conclude that GnRH neurons and fibers form a network within the anterior and medio-basal hypothalamus of llamas, suggesting the central location of mechanisms controlling reproductive processes in llamas (i.e., induced ovulation).
Collapse
Affiliation(s)
- Rodrigo A Carrasco
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada.
| | - Jaswant Singh
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada.
| | - Gregg P Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4, Canada.
| |
Collapse
|
105
|
Tan CL, Sheard PW, Jasoni CL. Developing neurites from mouse basal forebrain gonadotropin-releasing hormone neurons use Sonic hedgehog to modulate their growth. Int J Dev Neurosci 2018; 68:89-97. [PMID: 29787797 DOI: 10.1016/j.ijdevneu.2018.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/14/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons are required for fertility in all mammalian species studied to date. GnRH neuron cell bodies reside in the basal forebrain, and most extend long neurites in the caudal direction to terminate at the median eminence (ME), the site of hormone secretion. Using in vitro neurite growth assays, histological methods, and genetic deletion strategies in mice we have analysed the role of the morphogen and neurite growth and guidance molecule, Sonic hedgehog (Shh), in the growth of GnRH neurites to their target. Immunohistochemistry revealed that Shh was present in the basal forebrain, the preoptic area (POA) and mediobasal hypothalamus (MBH) at gestational day 14.5 (GD 14.5), a time when GnRH neurites grow towards the ME. Furthermore, in situ hybridization revealed that mRNA encoding the Shh receptor, Smoothened (Smo), was present in GnRH neurons from GD 15.5, when the first GnRH neurites are extending towards the MBH. In vitro neurite growth assays using hypothalamic explants from GD 15.5 fetuses in 3-D collagen gels showed that Shh was able to significantly stimulate GnRH neurite outgrowth. Finally, genetic deletion of Smo specifically from GnRH neurons in vivo, using Cre-loxP technology, resulted in a significant decrease in GnRH neurites innervating the ME. These experiments demonstrate that GnRH neurites use Shh for their neurite development, provide further understanding of the mechanisms by which GnRH nerve terminals arrive at their site of hormone secretion, and identify an additional hypothalamic neuronal population for which Shh/Smo signaling is developmentally important.
Collapse
Affiliation(s)
- C L Tan
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - P W Sheard
- Department of Physiology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| | - C L Jasoni
- Department of Anatomy, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand; Centre for Neuroendocrinology, University of Otago, School of Biomedical Sciences, Dunedin, 9054, New Zealand.
| |
Collapse
|
106
|
Brambilla C, Escobedo A, Artioli R, Lechuga MJ, Motta M, Bonadonna G. Medical Castration with Zoladex: A Conservative Approach to Premenopausal Breast Cancer. TUMORI JOURNAL 2018; 77:145-50. [PMID: 1828640 DOI: 10.1177/030089169107700211] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
For almost a century surgical castration represented the initial standard therapy for metastatic breast cancer in premenopausal women with hormone dependent tumors. Today the suppression of ovarian function can also be obtained by the administration of supraphysiologic doses of luteinizing hormone releasing hormone (LHRH) agonists. From April 1987 to February 1989, 23 premenopausal patients with advanced breast cancer (median age 39 years, range 28-52, ER positive 20, unknown 3; prior chemotherapy 17) were treated with the LHRH agonist goserelin depot (Zoladex) at the dose of 3.6 mg. every 4 weeks. Twenty-two patients were evaluable. Serum levels of 17 β estradiol, progesterone, FSH and LH were suppressed by goserelin and fell to postmenopausal values within 8 weeks of therapy in 77 % of cases. Complete response (CR) plus partial response (PR) was documented in 7 of 22 (32 %) and occurred in all major sites of disease. Five patients achieved CR (soft tissue 3, viscera 2). Response rate was higher in patients not previously treated with chemotherapy (4/6). In the present series, all responses were seen in women > 35 years old, regularly menstruating at the start of treatment. Time to progression for the entire case series was 22 weeks and for responders 64 weeks. Oophorectomy was performed after disease progression in four patients without success. Goserelin was well tolerated. Local cutaneous dyschromia occurred in 45 % and hot flushes in 82 %. Treatment efficacy of goserelin is comparable to that of oophorectomy, without the psychological trauma and the morbidity related to surgical castration.
Collapse
Affiliation(s)
- C Brambilla
- Division of Medical Oncology, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | |
Collapse
|
107
|
Chang JP, Pemberton JG. Comparative aspects of GnRH-Stimulated signal transduction in the vertebrate pituitary - Contributions from teleost model systems. Mol Cell Endocrinol 2018; 463:142-167. [PMID: 28587765 DOI: 10.1016/j.mce.2017.06.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/31/2017] [Accepted: 06/01/2017] [Indexed: 02/07/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) is a major regulator of reproduction through actions on pituitary gonadotropin release and synthesis. Although it is often thought that pituitary cells are exposed to only one GnRH, multiple GnRH forms are delivered to the pituitary of teleost fishes; interestingly this can include the cGnRH-II form usually thought to be non-hypophysiotropic. GnRHs can regulate other pituitary cell-types, both directly as well as indirectly, and multiple GnRH receptors (GnRHRs) may also be expressed in the pituitary, and even within a single pituitary cell-type. Literature on the differential actions of native GnRH isoforms in primary pituitary cells is largely derived from teleost fishes. This review will outline the diversity and complexity of GnRH-GnRHR signal transduction found within vertebrate gonadotropes as well as extra-gonadotropic sites with special emphasis on comparative studies from fish models. The implications that GnRHR transduction mechanisms are GnRH isoform-, function-, and cell-specific are also discussed.
Collapse
Affiliation(s)
- John P Chang
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | - Joshua G Pemberton
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
108
|
Faykoo-Martinez M, Monks DA, Zovkic IB, Holmes MM. Sex- and brain region-specific patterns of gene expression associated with socially-mediated puberty in a eusocial mammal. PLoS One 2018; 13:e0193417. [PMID: 29474488 PMCID: PMC5825099 DOI: 10.1371/journal.pone.0193417] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
The social environment can alter pubertal timing through neuroendocrine mechanisms that are not fully understood; it is thought that stress hormones (e.g., glucocorticoids or corticotropin-releasing hormone) influence the hypothalamic-pituitary-gonadal axis to inhibit puberty. Here, we use the eusocial naked mole-rat, a unique species in which social interactions in a colony (i.e. dominance of a breeding female) suppress puberty in subordinate animals. Removing subordinate naked mole-rats from this social context initiates puberty, allowing for experimental control of pubertal timing. The present study quantified gene expression for reproduction- and stress-relevant genes acting upstream of gonadotropin-releasing hormone in brain regions with reproductive and social functions in pre-pubertal, post-pubertal, and opposite sex-paired animals (which are in various stages of pubertal transition). Results indicate sex differences in patterns of neural gene expression. Known functions of genes in brain suggest stress as a key contributing factor in regulating male pubertal delay. Network analysis implicates neurokinin B (Tac3) in the arcuate nucleus of the hypothalamus as a key node in this pathway. Results also suggest an unappreciated role for the nucleus accumbens in regulating puberty.
Collapse
Affiliation(s)
| | - D. Ashley Monks
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Iva B. Zovkic
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
| | - Melissa M. Holmes
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto Mississauga, Mississauga, ON, Canada
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
109
|
Abstract
Neuropeptides are a diverse class of neuronal signalling molecules that regulate physiological processes and behaviour in animals. However, determining the relationships and evolutionary origins of the heterogeneous assemblage of neuropeptides identified in a range of phyla has presented a huge challenge for comparative physiologists. Here, we review revolutionary insights into the evolution of neuropeptide signalling that have been obtained recently through comparative analysis of genome/transcriptome sequence data and by ‘deorphanisation’ of neuropeptide receptors. The evolutionary origins of at least 30 neuropeptide signalling systems have been traced to the common ancestor of protostomes and deuterostomes. Furthermore, two rounds of genome duplication gave rise to an expanded repertoire of neuropeptide signalling systems in the vertebrate lineage, enabling neofunctionalisation and/or subfunctionalisation, but with lineage-specific gene loss and/or additional gene or genome duplications generating complex patterns in the phylogenetic distribution of paralogous neuropeptide signalling systems. We are entering a new era in neuropeptide research where it has become feasible to compare the physiological roles of orthologous and paralogous neuropeptides in a wide range of phyla. Moreover, the ambitious mission to reconstruct the evolution of neuropeptide function in the animal kingdom now represents a tangible challenge for the future. Summary: A review of the revolutionary advances in our knowledge of the evolution of neuropeptide signalling systems that have been enabled by comparative genomics and neuropeptide receptor deorphanisation.
Collapse
Affiliation(s)
- Maurice R Elphick
- School of Biological & Chemical Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Olivier Mirabeau
- Genetics and Biology of Cancers Unit, Institut Curie, INSERM U830, Paris Sciences et Lettres Research University, Paris 75005, France
| | - Dan Larhammar
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, 75124 Uppsala, Sweden
| |
Collapse
|
110
|
Abstract
Importance of the neuroendocrine brain for health and happiness has become clear since the 1960s. Foundations laid 100 years ago culminated in Geoffrey W Harris's model of control by the brain of secretion of anterior and posterior pituitary gland hormones through, respectively, releasing factors secreted into the hypothalamic-hypophysial portal system, and directly from axon terminals into the systemic circulation. Confirmation, expansion and deepening of knowledge and understanding have followed increasingly sophisticated technology. This allowed chemical characterisation of the posterior pituitary hormones, oxytocin and vasopressin, the releasing factors, their receptors and genes, location of the neurosecretory neurons in the hypothalamus, and how their activity is controlled, including by neural and hormonal feedback, and how hormone rhythms are generated. Wider roles of these neurons and their peptides in the brain are now recognised: in reproductive and social behaviours, emotions and appetite. Plasticity and epigenetic programming of neuroendocrine systems have emerged as important features.
Collapse
Affiliation(s)
- John A. Russell
- Professor Emeritus, Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, UK
| |
Collapse
|
111
|
Ullah R, Batool A, Wazir M, Naz R, Rahman TU, Wahab F, Shahab M, Fu J. Gonadotropin inhibitory hormone and RF9 stimulate hypothalamic-pituitary-adrenal axis in adult male rhesus monkeys. Neuropeptides 2017; 66:1-7. [PMID: 28757099 DOI: 10.1016/j.npep.2017.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 07/19/2017] [Accepted: 07/23/2017] [Indexed: 12/17/2022]
Abstract
Stress activates gonadotropin inhibitory hormone (GnIH), hypothalamic-pituitary-adrenal axis (HPA-axis) and represses hypothalamic-pituitary-gonadal axis (HPG-axis) but RF9 administration relieves stress-induced repression of the HPG-axis. Importantly, it was not known whether GnIH signaling and RF9 synthetic peptide modulate the HPA axis. To assess this, mammalian orthologs of GnIH (RFRP-1 and RFRP-3) and RF9 were administered to intact adult male rhesus monkeys. RFRP-1 (125μg/animal), RFRP-3 (250μg/animal) and RF9 (0.1mg/kg BW) were intravenously (iv) injected into normal fed (n=4) monkeys. Additionally, a single bolus iv injection of RF9 (0.1mg/kg BW) was also administered to 48h fasted monkeys (n=4) to check the effects of RF9 signaling on an activated HPA-axis. Serial blood samples were collected, centrifuged and the obtained plasma was used for the analysis of cortisol by specific enzyme immunoassay. RFRP-1 treatment significantly increased cortisol levels while RFRP-3 increased the plasma cortisol, but the effect was non-significant. RF9 treatment significantly increased cortisol levels in normal fed animals. In contrast, RF9 injection did not significantly alter circulating cortisol in fasted monkeys. In conclusion, our results suggest stimulatory action of RFRPs and RF9 on the HPA axis in the adult male monkeys. However, the mechanism and site of action of RFRP-1 and RF9 along the HPA-axis is still unknown. Therefore, further studies are needed to decipher the mechanism and site of action of RFRPs and RF9 on the HPA axis in primates.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou 310051, China; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Aalia Batool
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; The State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Chaoyang District, Beijing 100101, China
| | - Madiha Wazir
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Rabia Naz
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Tanzil Ur Rahman
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Department of Pathology and Pathophysiology, Key Laboratory of Reproductive Genetics, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Fazal Wahab
- Platform Degenerative Diseases, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, D-37077 Göttingen, Germany
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, Hangzhou 310051, China.
| |
Collapse
|
112
|
Banerjee S, Chaturvedi CM. Apoptotic mechanism behind the testicular atrophy in photorefractory and scotosensitive quail: Involvement of GnIH induced p-53 dependent Bax-Caspase-3 mediated pathway. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 176:124-135. [DOI: 10.1016/j.jphotobiol.2017.09.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 09/25/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022]
|
113
|
Testicular atrophy and reproductive quiescence in photorefractory and scotosensitive quail: Involvement of hypothalamic deep brain photoreceptors and GnRH-GnIH system. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2017; 175:254-268. [DOI: 10.1016/j.jphotobiol.2017.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 02/04/2023]
|
114
|
Perumal P, Savino N, Sangma CTR, Chang S, Sangtam TZT, Khan MH, Singh G, Kumar B, Yadav D, Srivastava N. Effect of season and age on scrotal circumference, testicular parameters and endocrinological profiles in mithun bulls. Theriogenology 2017; 98:23-29. [PMID: 28601151 DOI: 10.1016/j.theriogenology.2017.04.049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 04/29/2017] [Accepted: 04/29/2017] [Indexed: 10/19/2022]
Abstract
Mithun is a domesticated free-range bovine species primarily used as a meat animal and is a pride of North Eastern Hilly regions of India. The present study was conducted to measure the effect of seasons on scrotal & testicular biometrics and endocrinological profiles for different age groups at different seasons in mithun bulls. A total of 30 mithun males were selected from the mithun breeding farm, ICAR-NRC on Mithun, Medziphema, Nagaland, India and were equally distributed into five groups based on their age. Each group consisted of six animals and the groups were Gr A (0.1-1.0 year), Gr B (1.1-2.0 years), Gr C (2.1-3.0 years), Gr D (3.1-5.0 years) and Gr E (5.1-6.0 years). The seasons were grouped into winter, spring, summer and autumn based on the meteorological data such as temperature humidity index (THI) and sunshine hours. Scrotal circumference (SC) & testicular biometrics and endocrinological profiles such as follicle stimulating hormone (FSH), luteinizing hormone/interstitial cell stimulating hormone (LH/ICSH), testosterone, thyroxine (T4), cortisol and insulin like growth factor 1 (IGF 1) were estimated during different seasons for different age groups. Statistical results revealed that the SC & testicular biometrics and endocrinological profiles differed significantly (p < 0.05) among the different age groups for the same season whereas SC and endocrinological profiles significantly (p < 0.05) differed among the seasons for same age group. Significantly (p < 0.05) greater SC and testicular weight were observed in Gr E and D, lower in Gr A and B and intermediate in Gr C and increased as age advances. Significantly (p < 0.05) greater SC was observed in winter and spring and lowest was in summer season. The hormone profiles such as FSH, LH/ICSH, testosterone & thyroxine were significantly (p < 0.05) greater and IGF-1 & cortisol were significantly (p < 0.05) lower in spring & winter than in summer season. The hormones, FSH, LH/ICSH and thyroxine increased significantly in Gr A followed by reduced in Gr B and then increased to Gr D and E, whereas concentration of testosterone, cortisol and IGF-1 increased according to age advanced. It was concluded that the spring and winter seasons has significantly greater beneficial effects than summer season on reproduction and artificial breeding programme in semi-intensive management of mithun in the present location.
Collapse
Affiliation(s)
- P Perumal
- ICAR- National Research Centre on Mithun, Jharnapani, Nagaland, 797 106, India.
| | - N Savino
- NU-School of Agricultural Science and Rural Development, Medziphema, Nagaland, 797106, India
| | - C T R Sangma
- NU-School of Agricultural Science and Rural Development, Medziphema, Nagaland, 797106, India
| | - S Chang
- ICAR- National Research Centre on Mithun, Jharnapani, Nagaland, 797 106, India
| | - T Z T Sangtam
- NU-School of Agricultural Science and Rural Development, Medziphema, Nagaland, 797106, India
| | - M H Khan
- ICAR- National Research Centre on Mithun, Jharnapani, Nagaland, 797 106, India
| | - G Singh
- ICAR- Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243122, India
| | - Brijesh Kumar
- ICAR- ICAR Research Complex for NEH Region, Sikkim Centre, Sikkim, 737 103, India
| | - D Yadav
- ICAR- Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh, 243122, India
| | - N Srivastava
- ICAR-Central Institute for Research on Cattle, Meerut, 250001, India
| |
Collapse
|
115
|
Russell N, Cheung A, Grossmann M. Estradiol for the mitigation of adverse effects of androgen deprivation therapy. Endocr Relat Cancer 2017; 24:R297-R313. [PMID: 28667081 DOI: 10.1530/erc-17-0153] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 02/01/2023]
Abstract
Prostate cancer (PCa) is the second most commonly diagnosed cancer in men. Conventional endocrine treatment for PCa leads to global sex steroid deprivation. The ensuing severe hypogonadism is associated with well-documented adverse effects. Recently, it has become apparent that many of the biological actions attributed to androgens in men are in fact not direct, but mediated by estradiol. Available evidence supports a primary role for estradiol in vasomotor stability, skeletal maturation and maintenance, and prevention of fat accumulation. Hence there has been interest in revisiting estradiol as a treatment for PCa. Potential roles for estradiol could be in lieu of conventional androgen deprivation therapy or as low-dose add-back treatment while continuing androgen deprivation therapy. These strategies may limit some of the side effects associated with conventional androgen deprivation therapy. However, although available data are reassuring, the potential for cardiovascular risk and pro-carcinogenic effects on PCa via estrogen receptor signalling must be considered.
Collapse
Affiliation(s)
- Nicholas Russell
- Department of EndocrinologyAustin Health, Heidelberg, Victoria, Australia
- Department of Medicine (Austin Health)The University of Melbourne, Heidelberg, Victoria, Australia
| | - Ada Cheung
- Department of EndocrinologyAustin Health, Heidelberg, Victoria, Australia
- Department of Medicine (Austin Health)The University of Melbourne, Heidelberg, Victoria, Australia
| | - Mathis Grossmann
- Department of EndocrinologyAustin Health, Heidelberg, Victoria, Australia
- Department of Medicine (Austin Health)The University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
116
|
Dubovy SR, Fernandez MP, Echegaray JJ, Block NL, Unoki N, Perez R, Vidaurre I, Lee RK, Nadji M, Schally AV. Expression of hypothalamic neurohormones and their receptors in the human eye. Oncotarget 2017; 8:66796-66814. [PMID: 28977997 PMCID: PMC5620137 DOI: 10.18632/oncotarget.18358] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/20/2017] [Indexed: 12/27/2022] Open
Abstract
Extrapituitary roles for hypothalamic neurohormones have recently become apparent and clinically relevant, based on the use of synthetic peptide analogs for the treatment of multiple conditions including cancers, pulmonary edema and myocardial infarction. In the eye, it has been suggested that some of these hormones and their receptors may be present in the ciliary body, iris, trabecular meshwork and retina, but their physiological role has yet to be elucidated. Our study intends to comprehensively demonstrate the expression of some hypothalamic neuroendocrine hormones and their receptors within different retinal and extraretinal structures of the human eye. Immunofluorescence, Western blot analysis, and RT-PCR were used to evaluate the qualitative and quantitative expression of Luteinizing Hormone Releasing Hormone (LHRH), Growth Hormone Releasing Hormone (GHRH), Thyrotropin Releasing Hormone (TRH), Gastrin Releasing Peptide (GRP) and Somatostatin as well as their respective receptors (LHRH-R, GHRH-R, TRH-R, GRP-R, SST-R1) in cadaveric human eye tissue and in paraffinized human eye tissue sections. The hypothalamic hormones LHRH, GHRH, TRH, GRP and Somatostatin and their respective receptors (LHRH-R, GHRH-R, TRH-R, GRPR/BB2 and SST-R1), were expressed in the conjunctiva, cornea, trabecular meshwork, ciliary body, lens, retina, and optic nerve.
Collapse
Affiliation(s)
- Sander R Dubovy
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Maria P Fernandez
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Jose J Echegaray
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Department of Ophthalmology, University of Puerto Rico School of Medicine, San Juan, PR, USA
| | - Norman L Block
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Noriyuki Unoki
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Roberto Perez
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | | | - Richard K Lee
- Florida Lions Ocular Pathology Laboratory, Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Mehrdad Nadji
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA
| | - Andrew V Schally
- Miami Veterans Affairs Medical Center, Miami, Florida, USA.,Department of Pathology, University of Miami, Miller School of Medicine, Miami, Florida, USA.,Divisions of Hematology/Oncology, Endocrinology, Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
117
|
Schally AV, Block NL, Rick FG. Discovery of LHRH and development of LHRH analogs for prostate cancer treatment. Prostate 2017; 77:1036-1054. [PMID: 28449236 DOI: 10.1002/pros.23360] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/06/2023]
Abstract
The discovery, isolation, elucidation of structure, synthesis, and initial testing of the neuropeptide hypothalamic luteinizing hormone-releasing hormone (LHRH), which regulates reproduction, is briefly described. The design, synthesis, and experimental and clinical testing of agonistic analogs of LHRH is extensively reviewed focusing on the development of new methods for the treatment of prostate cancer. Subsequent development of antagonistic analogs of LHRH is then faithfully recounted with special emphasis on therapy of prostate cancer and BPH. The concepts of targeted therapy to peptide receptors on tumors are re-examined and the development of the cytotoxic analogs of LHRH and their status is reviewed. The endeavor to develop better therapies for prostate cancer, based on LHRH analogs, guided much of our work.
Collapse
Affiliation(s)
- Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Division of Endocrinology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Norman L Block
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida
- Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
118
|
Ullah R, Su Y, Shen Y, Li C, Xu X, Zhang J, Huang K, Rauf N, He Y, Cheng J, Qin H, Zhou YD, Fu J. Postnatal feeding with high-fat diet induces obesity and precocious puberty in C57BL/6J mouse pups: a novel model of obesity and puberty. Front Med 2017; 11:266-276. [DOI: 10.1007/s11684-017-0530-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/08/2017] [Indexed: 12/15/2022]
|
119
|
Ng J, Chwalisz K, Carter DC, Klein CE. Dose-Dependent Suppression of Gonadotropins and Ovarian Hormones by Elagolix in Healthy Premenopausal Women. J Clin Endocrinol Metab 2017; 102:1683-1691. [PMID: 28323948 DOI: 10.1210/jc.2016-3845] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 02/13/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT Elagolix is a nonpeptide, oral gonadotropin-releasing hormone (GnRH) antagonist being developed for sex-hormone-dependent diseases in women. OBJECTIVE We evaluated the pharmacokinetics and pharmacodynamics of elagolix. DESIGN, SETTING, AND PARTICIPANTS This study was a randomized, double-blind, placebo-controlled, multiple-ascending dose study in 45 healthy premenopausal women at a research unit. INTERVENTIONS Elagolix [150 mg once daily or 100, 200, 300, or 400 mg twice daily (BID)] or placebo was administered for 21 days. MAIN OUTCOME MEASURES Main outcome measures were elagolix pharmacokinetics, suppression of gonadotropics [follicle-stimulating hormone (FSH), luteinizing hormone (LH)] and ovarian hormones [estradiol (E2), progesterone (P)], and adverse events. RESULTS Elagolix was rapidly absorbed after oral dosing, reaching maximum concentrations at 1.0 to 1.5 hours, with a half-life of 4 to 6 hours. FSH, LH, and E2 were suppressed within hours of elagolix administration on day 1. Dose-dependent suppression of E2 was observed, with maximum suppression achieved with elagolix 200 mg BID. Dose-dependent suppression of FSH and LH was also observed, with maximal or near-maximal suppression achieved at 300 mg BID and 200 mg BID, respectively. At elagolix doses ≥100 mg BID, P concentrations remained at anovulatory levels throughout 21 days of dosing. The most frequently reported adverse events were headache and hot flush. CONCLUSIONS Elagolix administration allows for modulation of gonadotropin and ovarian hormone concentrations, from partial suppression at lower doses to nearly full suppression at higher doses. The results of this study provide a rationale for elagolix dose selection for treatment of sex hormone-dependent diseases in women.
Collapse
Affiliation(s)
- Juki Ng
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois 60064
| | | | - David C Carter
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois 60064
| | - Cheri E Klein
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., North Chicago, Illinois 60064
| |
Collapse
|
120
|
Interleukin-18 and its receptor are expressed in gonadotropin-releasing hormone neurons of mouse and rat forebrain. Neurosci Lett 2017; 650:33-37. [DOI: 10.1016/j.neulet.2017.03.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/20/2017] [Accepted: 03/31/2017] [Indexed: 11/19/2022]
|
121
|
Characterization and spatiotemporal expression of gonadotropin-releasing hormone in the Pacific abalone, Haliotis discus hannai. Comp Biochem Physiol A Mol Integr Physiol 2017; 209:1-9. [PMID: 28408352 DOI: 10.1016/j.cbpa.2017.04.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/23/2017] [Accepted: 04/03/2017] [Indexed: 11/20/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a key neuropeptide regulating reproduction in humans and other vertebrates. Recently, GnRH-like cDNAs and peptides were reported in marine mollusks, implying that GnRH-mediated reproduction is an ancient neuroendocrine system that arose prior to the divergence of protostomes and deuterostomes. Here, we evaluated the reproductive control system mediated by GnRH in the Pacific abalone Haliotis discus hannai. We cloned a prepro-GnRH cDNA (Hdh-GnRH) from the pleural-pedal ganglion (PPG) in H. discus hannai, and analyzed its spatiotemporal gene expression pattern. The open reading frame of Hdh-GnRH encodes a protein of 101 amino acids, consisting of a signal peptide, a GnRH dodecapeptide, a cleavage site, and a GnRH-associated peptide. This structure and sequence are highly similar to GnRH-like peptides reported for mollusks and other invertebrates. Quantitative polymerase chain reaction demonstrated that Hdh-GnRH mRNA was more strongly expressed in the ganglions (PPG and cerebral ganglion [CG]) than in other tissues (gonads, gills, intestine, hemocytes, muscle, and mantle) in both sexes. In females, the expression levels of Hdh-GnRH mRNA in the PPG and branchial ganglion (BG) were significantly higher at the ripe and partial spent stages than at the early and late active stages. In males, Hdh-GnRH mRNA levels in the BG showed a significant increase in the partial spent stage. Unexpectedly, Hdh-GnRH levels in the CG were not significantly different among the examined stages in both sexes. These results suggest that Hdh-GnRH mRNA expression profiles in the BG and possibly the PPG are tightly correlated with abalone reproductive activities.
Collapse
|
122
|
Horton KW, Carlson NE, Grunwald GK, Mulvahill MJ, Polotsky AJ. A population-based approach to analyzing pulses in time series of hormone data. Stat Med 2017; 36:2576-2589. [PMID: 28393373 DOI: 10.1002/sim.7292] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 02/23/2017] [Accepted: 03/01/2017] [Indexed: 01/28/2023]
Abstract
Studies of reproductive physiology involve rapid sampling protocols that result in time series of hormone concentrations. The signature pattern in these times series is pulses of hormone release. Various statistical models for quantifying the pulsatile release features exist. Currently these models are fitted separately to each individual and the resulting estimates averaged to arrive at post hoc population-level estimates. When the signal-to-noise ratio is small or the time of observation is short (e.g., 6 h), this two-stage estimation approach can fail. This work extends the single-subject modelling framework to a population framework similar to what exists for complex pharamacokinetics data. The goal is to leverage information across subjects to more clearly identify pulse locations and improve estimation of other model parameters. This modelling extension has proven difficult because the pulse number and locations are unknown. Here, we show that simultaneously modelling a group of subjects is computationally feasible in a Bayesian framework using a birth-death Markov chain Monte Carlo estimation algorithm. Via simulation, we show that this population-based approach reduces the false positive and negative pulse detection rates and results in less biased estimates of population-level parameters of frequency, pulse size, and hormone elimination. We then apply the approach to a reproductive study in healthy women where approximately one-third of the 21 subjects in the study did not have appropriate fits using the single-subject fitting approach. Using the population model produced more precise, biologically plausible estimates of all model parameters. Copyright © 2017 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- K W Horton
- HQ USAFA/DFMS, 2354 Fairchild Dr, USAF Academy, 80840, CO, U.S.A
| | - N E Carlson
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, 13001 E. 17th PL, MS B119, Aurora, 80045, CO, U.S.A
| | - G K Grunwald
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, 13001 E. 17th PL, MS B119, Aurora, 80045, CO, U.S.A
| | - M J Mulvahill
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, 13001 E. 17th PL, Aurora, 80045, CO, U.S.A
| | - A J Polotsky
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, MS B198-3, Aurora, 80045, CO, U.S.A
| |
Collapse
|
123
|
Park WC. Role of Ovarian Function Suppression in Premenopausal Women with Early Breast Cancer. J Breast Cancer 2016; 19:341-348. [PMID: 28053622 PMCID: PMC5204040 DOI: 10.4048/jbc.2016.19.4.341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/07/2016] [Indexed: 01/21/2023] Open
Abstract
Historically, endocrine therapy for breast cancer began with ovarian ablation (OA) for the treatment of premenopausal patients. After the identification of estrogen receptors and the development of many antiestrogens, tamoxifen has been approved and used as the standard endocrine therapy for hormonal receptor (HR)-positive premenopausal patients to date. With the development of luteinizing hormone-releasing hormone agonists, the paradigm of endocrine therapy for premenopausal women with HR-positive breast cancer began to change from OA to ovarian function suppression (OFS). To date, the indication for OFS was limited to those premenopausal patients with HR-positive breast cancer who were unable to use tamoxifen as the primary adjuvant endocrine therapy. However, following the definitive demonstration of the therapeutic role of OFS added to tamoxifen or aromatase inhibitor after chemotherapy in large randomized trials, such as Tamoxifen and Exemestane Trial or Suppression of Ovarian Function Trial, the American Society of Clinical Oncology guidelines for the use of endocrine therapy in premenopausal HR-positive breast cancer were recently updated to recommend OFS in high-risk patients who required adjuvant chemotherapy. In contrast, the role of OFS to protect ovarian function during chemotherapy in premenopausal women has remained controversial, and some evidence showing the protective effect of OFS on the ovaries during chemotherapy as well as its therapeutic effect for breast cancer in premenopausal women with HR-negative breast cancer was recently published. Further evaluation is necessary to determine its exact role. In conclusion, the role of OA or OFS has been evolving, not only to improve the efficacy of breast cancer treatment, but also to preserve ovary function. OFS remains a main strategy for premenopausal women with HR-positive early breast cancer, though its exact role should be determined in further studies.
Collapse
Affiliation(s)
- Woo-Chan Park
- Department of Surgery, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
124
|
Hoda MR, Kramer MW, Merseburger AS, Cronauer MV. Androgen deprivation therapy with Leuprolide acetate for treatment of advanced prostate cancer. Expert Opin Pharmacother 2016; 18:105-113. [PMID: 27826989 DOI: 10.1080/14656566.2016.1258058] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Hormone sensitive advanced prostate cancer (PCa) is an incurable disease that is treated with a variety of hormonal therapies targeting the androgen/androgen receptor signaling axis. For decades androgen deprivation therapy (ADT) by surgical or chemical castration is the gold standard for the treatment of advanced PCa. Areas covered: This review discusses the pharmacological features of Leuprolide, a luteinizing hormone-releasing hormone (LHRH) agonists/analog and the most commonly used drug in ADT. Expert opinion: Although Leuprolide has been on the market for more than 30 years it is still the leading option for ADT and serves as a basis for most multimodal therapy concepts. The fact that with the onset of castration-resistance in late stage metastatic disease, a prolongation of ADT in combination with a second line hormonal manipulation is recommended supports the importance of the compound for daily clinical practice.
Collapse
Affiliation(s)
- M Raschid Hoda
- a Department of Urology , University Clinic of Schleswig-Holstein , Lübeck , Germany
| | - Mario W Kramer
- a Department of Urology , University Clinic of Schleswig-Holstein , Lübeck , Germany
| | - Axel S Merseburger
- a Department of Urology , University Clinic of Schleswig-Holstein , Lübeck , Germany
| | - Marcus V Cronauer
- a Department of Urology , University Clinic of Schleswig-Holstein , Lübeck , Germany
| |
Collapse
|
125
|
Calisi RM, Geraghty AC, Avila A, Kaufer D, Bentley GE, Wingfield JC. Patterns of hypothalamic GnIH change over the reproductive period in starlings and rats. Gen Comp Endocrinol 2016; 237:140-146. [PMID: 27591072 DOI: 10.1016/j.ygcen.2016.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/05/2016] [Accepted: 08/24/2016] [Indexed: 11/30/2022]
Abstract
Gonadotropin inhibitory hormone (GnIH) exerts powerful inhibitory effects on various levels of the vertebrate hypothalamic-pituitary-gonadal (reproductive) axis, yet little is known of how it might change naturally over the course of reproduction. We characterized patterns of hypothalamic GnIH cell abundance over the reproductive period in two popular models used for the study of reproductive endocrinology: European starlings (Sturnus vulgaris) and Sprague-Dawley rats (Rattus norvegicus). We also examined the effects on an unpredictable change in the environment on GnIH cell abundance during the reproductive period, specifically during the period of parental care, by simulating a nest predation event and removing eggs/pups. In both species, we report changes in GnIH cell abundance are occurring at similar reproductive time points but are not always directionally parallel; this may be due to a difference in life histories and physiology mediating parental care. We discovered that cells immunoreactive for the GnIH peptide in male and female starlings are most highly abundant on the first day of incubation and the first day after the first chick hatches. Conversely in rats, GnIH cell abundance decreases in dams on the first day after pups are born. In both male and female starlings and female rats, GnIH cell abundance increases in response to egg/pup loss, indicating that GnIH responds to an unpredictable change in the environment in a potentially conserved fashion. These changes in GnIH cell abundance during the reproductive period inspire further investigation of its adaptive role in reproductive physiological events and behaviors, especially parental care.
Collapse
Affiliation(s)
- R M Calisi
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, USA; Department of Integrative Biology, University of California, Berkeley, CA, USA.
| | - A C Geraghty
- Department of Integrative Biology, University of California, Berkeley, CA, USA
| | - A Avila
- Department of Integrative Biology, University of California, Berkeley, CA, USA
| | - D Kaufer
- Department of Integrative Biology, University of California, Berkeley, CA, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - G E Bentley
- Department of Integrative Biology, University of California, Berkeley, CA, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| | - J C Wingfield
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA, USA
| |
Collapse
|
126
|
Ullah R, Shen Y, Zhou YD, Huang K, Fu JF, Wahab F, Shahab M. Expression and actions of GnIH and its orthologs in vertebrates: Current status and advanced knowledge. Neuropeptides 2016; 59:9-20. [PMID: 27255391 DOI: 10.1016/j.npep.2016.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/01/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022]
Abstract
The physiology of reproduction is very complex and is regulated by multiple factors, including a number of hypothalamic neuropeptides. In last few decades, various neuropeptides have been discovered to be involved in stimulation or inhibition of reproduction. In 2000, Tsutsui and colleagues uncovered gonadotropin-inhibitory hormone (GnIH), a neuropeptide generating inhibitory drive to the reproductive axis, in the brain of Coturnix quail. Afterward, GnIH orthologs were discovered in other vertebrates from fish to mammals including human. In these vertebrates, all the discovered GnIH and its ortholgs have LPXRFamide (X=L or Q) sequence at C-terminus. GnIH orthologs of mammals and primates are also termed as RFamide-related peptide (RFRP)-1 and -3 that too have an LPXRFamide (X=L or Q) motif at their C-terminus. GnIH and its orthologs form a member of the RFamide peptide family. GnIH signals via its canonical G protein coupled receptor 147 (GPR147). Both GnIH and GPR147 are expressed in hypothalamus and other brain regions. Besides actions through the hypothalamic GnRH and kisspeptinergic neurons, GnIH-GPR147 signaling exerts inhibitory effect on the reproductive axis via pituitary gonadotropes and directly at gonadal level. Various factors including availability and quality of food, photoperiod, temperature, social interaction, various stresses and some diseases modulate GnIH-GPR147 signaling. In this review, we have discussed expression and actions of GnIH and its orthologs in vertebrates. Special emphasis is given on the role of GnIH-GPR147 signaling pathway in the regulation of reproduction. We have also reviewed and discussed currently available literature on the participation of GnIH-GPR147 signaling pathway in the stress modulation of reproduction.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Yi Shen
- Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Yu-Dong Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health and Zhejiang Province, Department of Neurobiology, School of Medicine, Zhejiang University, Hangzhou 310058, PR China
| | - Ke Huang
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan; Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jun-Fen Fu
- Department of Endocrinology, Children Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Fazal Wahab
- Stem Cell Biology Unit, German Primate Center, Leibniz-Institute for Primate Research, Kellnerweg 4, 37077 Gottingen, Germany
| | - Muhammad Shahab
- Laboratory of Reproductive Neuroendocrinology, Department of Animal Sciences, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
127
|
Miccoli A, Olivotto I, De Felice A, Leonori I, Carnevali O. Characterization and transcriptional profiles of Engraulis encrasicolus' GnRH forms. Reproduction 2016; 152:727-739. [PMID: 27651520 DOI: 10.1530/rep-16-0405] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/19/2016] [Indexed: 12/12/2022]
Abstract
The European anchovy Engraulis encrasicolus, a member of the Clupeiformes order, holds a great biological and economical importance. In the past, this species was mostly investigated with the aim of assessing its reproductive biology, trophic ecology, population dynamics and the relations existing with the physical environment. At present days, though, an almost complete lack of information afflicts its neuroendocrinology and reproductive physiology. The hypothalamic-pituitary-gonadal (HPG) axis at its highest levels was herein investigated. In this study, the gonadotropin-releasing hormone (GnRH), a neuropeptide underlying many reproduction-related processes, the most critical of which is the stimulation of gonadotropin synthesis and secretion from the pituitary gland, was cloned. Three forms (salmon GnRH, chicken-II GnRH and the species-specific type) were characterized in their full-length open-reading frames and, in accordance with other Clupeiformes species, the distinctive one was found to be the herring-type GnRH. We qualitatively and semiquantitatively evaluated the localizations of expressions and the temporal transcription patterns of the three GnRH forms in male and female specimens throughout their reproductive cycle as well as described their phylogeny with regard to teleost GnRH lineages, and, specifically, to other Clupeiformes species.
Collapse
Affiliation(s)
- Andrea Miccoli
- Department of Life and Environmental SciencesUniversità Politecnica delle Marche, Ancona, Italy.,CNR-National Research Council of ItalyISMAR-Marine Sciences Institute, Ancona, Italy
| | - Ike Olivotto
- Department of Life and Environmental SciencesUniversità Politecnica delle Marche, Ancona, Italy
| | - Andrea De Felice
- CNR-National Research Council of ItalyISMAR-Marine Sciences Institute, Ancona, Italy
| | - Iole Leonori
- CNR-National Research Council of ItalyISMAR-Marine Sciences Institute, Ancona, Italy
| | - Oliana Carnevali
- Department of Life and Environmental SciencesUniversità Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
128
|
Uenoyama Y, Pheng V, Tsukamura H, Maeda KI. The roles of kisspeptin revisited: inside and outside the hypothalamus. J Reprod Dev 2016; 62:537-545. [PMID: 27478063 PMCID: PMC5177970 DOI: 10.1262/jrd.2016-083] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Kisspeptin, encoded by KISS1/Kiss1 gene, is now considered a master regulator of reproductive functions in mammals owing to
its involvement in the direct activation of gonadotropin-releasing hormone (GnRH) neurons after binding to its cognate receptor, GPR54. Ever since the discovery
of kisspeptin, intensive studies on hypothalamic expression of KISS1/Kiss1 and on physiological roles of hypothalamic
kisspeptin neurons have provided clues as to how the brain controls sexual maturation at the onset of puberty and subsequent reproductive performance in
mammals. Additionally, emerging evidence indicates the potential involvement of extra-hypothalamic kisspeptin in reproductive functions. Here, we summarize data
regarding kisspeptin inside and outside the hypothalamus and revisit the physiological roles of central and peripheral kisspeptins in the reproductive functions
of mammals.
Collapse
Affiliation(s)
- Yoshihisa Uenoyama
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | | | | | | |
Collapse
|
129
|
Schally AV, Block NL, Rick FG. New therapies for relapsed castration-resistant prostate cancer based on peptide analogs of hypothalamic hormones. Asian J Androl 2015; 17:925-8. [PMID: 26112478 PMCID: PMC4814950 DOI: 10.4103/1008-682x.152819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is a pleasure to contribute our presentation at the International Prostate Forum of the Annual Meeting of the American Urological Association (AUA) to this special issue of the Asian Journal of Andrology.
Collapse
Affiliation(s)
- Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Endocrinology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Norman L Block
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Medicine, Division of Hematology/Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL 33125, USA
- Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33174, USA
| |
Collapse
|
130
|
Abstract
SUMMARYThe results of lesion, stimulation, deafferentation, implantatión and transplantation studies employed in the identification of hypophysiotrophic control areas in the hypothalamus to date suggest the following probable locations: corticotropic releasing factor (CRF) is formed in a diffuse area along the base of the median eminence, if not the base of the entire hypothalamus. Follicle stimulating hormone releasing factor (FSHRF) is elaborated in the paraventricular-suprachiasmatic areas but its cyclic control may reside in the anterior hypothalamic area. Luteinizing hormone (LH) is controlled by luteinizing hormone releasing factor (LHRF) formed in the suprachiasmatic area: its cyclic control may be in the preoptic area. Prolactin is controlled by prolactin inhibiting factor (PIF) localized in a diffuse area comprising the ventromedial, dorsomedial, arcuate and paraventricular nuclei. The hypothalamic area involved in thyroid control is also rather large, since thyrotropin stimulating hormone releasing factor (TSHRF) has been found in an area including the supraoptic and chiasmatic nuclei. Growth hormone releasing factor (GHRF) is elaborated in a rather narrow zone, the ventromedial hypothalamic nuclei.
Collapse
|
131
|
Vega MG, Zarek SM, Bhagwat M, Segars JH. Gonadotropin surge-inhibiting/attenuating factors: a review of current evidence, potential applications, and future directions for research. Mol Reprod Dev 2015; 82:2-16. [PMID: 25581424 DOI: 10.1002/mrd.22439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 10/29/2014] [Indexed: 11/12/2022]
Abstract
Animal studies in the 1980s suggested the existence of an ovarian hormone, termed gonadotropin surge-inhibiting/attenuating factor (GnSIF/AF), that modulates pituitary secretion of luteinizing hormone (LH). Given the importance of identifying regulatory factors of the hypothalamic-pituitary-ovarian axis and the accumulating data suggesting its existence, we conducted a comprehensive literature search using PubMed, Web of Science, Scopus, and Embase to identify articles related to GnSIF/AF. The search generated 161 publications, of which 97 were included in this study. Several attempts have been made to identify and characterize this hormone and several candidates have been identified, but the protein sequences of these putative GnSIF/AF factors differ widely from one study to another. In addition, while the RF-amide RFRP-3 is known foremost as a neuropeptide, some research supports an ovarian origin for this non-steroidal hormone, thereby suggesting a role for RFRP-3 either as a co-modulator of GnSIF/AF or as a gonadotropin-inhibiting factor in the hypothalamus (GnIH). Discovery of the KNDy neurons that modulate GnRH secretion, on the other hand, further encourages the search for substance(s) that modulate their activity and that indirectly affect LH secretion and the hypothalamic-pituitary-ovarian axis. While it has remained an elusive hormone, GnSIF/AF holds many potential applications for contraception, in vitro fertilization, and/or cancer as well as for understanding polycystic ovary syndrome, metabolic diseases, and/or pubertal development. In this review, we rigorously examine the available evidence regarding the existence of GnSIF/AF, previous attempts at its identification, limitations to its discovery, future directions of research, and potential clinical applications.
Collapse
Affiliation(s)
- Mario G Vega
- Department of Obstetrics and Gynecology, St. Luke's-Roosevelt Hospital, New York City, New York
| | | | | | | |
Collapse
|
132
|
Forni PE, Wray S. GnRH, anosmia and hypogonadotropic hypogonadism--where are we? Front Neuroendocrinol 2015; 36:165-77. [PMID: 25306902 PMCID: PMC4703044 DOI: 10.1016/j.yfrne.2014.09.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 09/08/2014] [Accepted: 09/25/2014] [Indexed: 11/19/2022]
Abstract
Gonadotropin releasing hormone (GnRH) neurons originate the nasal placode and migrate into the brain during prenatal development. Once within the brain, these cells become integral components of the hypothalamic-pituitary-gonadal axis, essential for reproductive function. Disruption of this system causes hypogonadotropic hypogonadism (HH). HH associated with anosmia is clinically defined as Kallman syndrome (KS). Recent work examining the developing nasal region has shed new light on cellular composition, cell interactions and molecular cues responsible for the development of this system in different species. This review discusses some developmental aspects, animal models and current advancements in our understanding of pathologies affecting GnRH. In addition we discuss how development of neural crest derivatives such as the glia of the olfactory system and craniofacial structures control GnRH development and reproductive function.
Collapse
Affiliation(s)
- Paolo E Forni
- Department of Biological Sciences and the Center for Neuroscience Research, University at Albany, State University of New York, Albany, NY 12222, United States.
| | - Susan Wray
- Cellular and Developmental Neurobiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
133
|
Rick FG, Schally AV. Bench-to-bedside development of agonists and antagonists of luteinizing hormone-releasing hormone for treatment of advanced prostate cancer. Urol Oncol 2014; 33:270-4. [PMID: 25512159 DOI: 10.1016/j.urolonc.2014.11.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/03/2014] [Accepted: 11/11/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Androgen deprivation therapy (ADT) has been the standard of care for treating patients with hormone-sensitive advanced prostate cancer (PCa) for 3 decades. The agonists of luteinizing hormone-releasing hormone (LHRH), also called gonadotropin-releasing hormone, are still the most frequently used form of medical ADT. ADT AND LHRH ANALOGS The application of agonists of LHRH has improved and modernized the treatment of advanced PCa; millions of patients have benefited from therapy with LHRH agonists as a preferred alternative to surgical castration, as the psychological effects and perpetuity of orchiectomy are undesirable for most men. Despite their efficacy, agonists of LHRH have several shortcomings, including initial surge in testosterone, producing exacerbation of clinical symptoms, and microsurges in testosterone that might occur after each administration. A new, alternate approach to ADT is emerging with the improvements in antagonists of LHRH. This class of LHRH analogues produces a direct and immediate blockade of pituitary LHRH receptors and leads to a more rapid suppression of testosterone without an initial surge or subsequent microsurges. Degarelix, a third-generation LHRH antagonist, is the only antagonist with a low histamine-releasing activity that is currently on the market for clinical use in advanced PCa with improved testosterone suppression, better control of follicle-stimulating hormone and prostate-specific antigen, and which offers a prolonged delay to progression and more favorable effects on serum alkaline phosphatase. CONCLUSIONS Although LHRH agonists are still the mainstay for treatment of advanced PCa, antagonists of LHRH offer an alternative as a pharmacological approach.
Collapse
Affiliation(s)
- Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL.
| | - Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL; Division of Hematology/Oncology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL; Division of Endocrinology, Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL
| |
Collapse
|
134
|
Lima CJG, Cardoso SC, Lemos EFL, Zingler E, Capanema C, Menezes LD, Vogado G, Dos Santos BTA, de Moraes OL, Duarte EF, de Brito VN, Latronico AC, Lofrano-Porto A. Mutational analysis of the genes encoding RFamide-related peptide-3, the human orthologue of gonadotrophin-inhibitory hormone, and its receptor (GPR147) in patients with gonadotrophin-releasing hormone-dependent pubertal disorders. J Neuroendocrinol 2014; 26:817-24. [PMID: 25180599 DOI: 10.1111/jne.12207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 08/24/2014] [Accepted: 08/27/2014] [Indexed: 11/26/2022]
Abstract
RFamide-related peptide-3 (RFRP-3), the orthologue of avian gonadotrophin-inhibitory hormone, and its receptor GPR147 have been recently identified in the human hypothalamus, and their roles in the regulation of reproductive axis has been studied. The present study aimed to investigate whether the presence of variants in the genes encoding human RFRP-3 (NPVF gene) and its receptor, GPR147 (NPFFR1 gene), is associated with the occurrence of gonadotrophin-releasing hormone-dependent pubertal disorders. Seventy-eight patients with idiopathic central precocious puberty (CPP) and 51 with normosmic isolated hypogonadotrophic hypogonadism (nIHH) were investigated. Fifty healthy subjects comprised the control group. The coding sequences of the NPVF and NPFFR1 genes were amplified and sequenced. Odds ratios (OR) were used to estimate the likelihood of CPP or nIHH in the presence of the described polymorphisms. All such polymorphisms have already been registered in the National Center for Biotechnology Information database. A three-nucleotide in frame deletion was identified in the NPVF gene (p.I71_K72), with a smaller proportion in the CPP (5%) compared to the nIHH (15%) group (P = 0.06). This results in the deletion of the isoleucine at position 71, adjacent to lysine at an endoproteolytic cleavage site of the precursor peptide. This polymorphism was associated with a lower risk of CPP (OR = 0.33; 95% confidence interval = 0.08-0.88); interestingly, only two men with nIHH were homozygotes for this variant. A total of five missense polymorphisms were found in the NPFFR1 gene, which encodes GPR147, with similar frequencies among groups and no association with pubertal timing. Our data suggest that RFRP-3/GPR147 may play secondary, modulatory roles on the regulation of pubertal development; a restraining modulatory effect of the NPVF p.I71_K72 variant on the activation of the gonadotrophic axis cannot be ruled out and deserves further investigation.
Collapse
Affiliation(s)
- C J G Lima
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde e Ambulatório de Endocrinologia das Gônadas e Adrenais, Hospital Universitário de Brasília, Universidade de Brasília, Brasília, DF, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
135
|
Chism DD, De Silva D, Whang YE. Mechanisms of acquired resistance to androgen receptor targeting drugs in castration-resistant prostate cancer. Expert Rev Anticancer Ther 2014; 14:1369-78. [PMID: 24927631 PMCID: PMC4221359 DOI: 10.1586/14737140.2014.928594] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
After initial response to androgen receptor (AR) targeting drugs abiraterone or enzalutamide, most patients develop progressive disease and therefore, castration resistant prostate cancer remains a terminal disease. Multiple mechanisms underlying acquired resistance have been postulated. Intratumoral androgen synthesis may resume after abiraterone treatment. A point mutation in the ligand-binding domain of AR may confer resistance to enzalutamide. Emergence of AR splice variants lacking the ligand-binding domain may mediate resistance to abiraterone and enzalutamide. Steroid receptors such as glucocorticoid receptor may substitute for AR. Drugs with novel mechanisms of action or combination therapy, along with biomarkers for patient selection, may be needed to improve the therapy of castration resistant prostate cancer.
Collapse
Affiliation(s)
- David D. Chism
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599
| | - Dinuka De Silva
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599
| | - Young E. Whang
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599
| |
Collapse
|
136
|
López-Doval S, Salgado R, Pereiro N, Moyano R, Lafuente A. Perfluorooctane sulfonate effects on the reproductive axis in adult male rats. ENVIRONMENTAL RESEARCH 2014; 134:158-168. [PMID: 25171141 DOI: 10.1016/j.envres.2014.07.006] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/08/2014] [Accepted: 07/12/2014] [Indexed: 06/03/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a neurotoxic agent and it can disrupt the endocrine system activity. This work was undertaken to evaluate the possible effects of PFOS exposure on the hypothalamic-pituitary-testicular axis (HPT) in adult male rats, and to evaluate the possible morphological alterations induced by PFOS in the endocrine tissues of this axis. Adult male rats were orally treated with 0.5; 1.0; 3.0 and 6.0 mg of PFOS/kg/day for 28 days. After PFOS exposure, hypothalamic noradrenaline concentration increased in the anterior hypothalamus and in the median eminence, not changing in the mediobasal hypothalamus. PFOS treated rats presented a decrease of the gonadotropin releasing hormone (GnRH) gene expression, increasing the mRNA levels of the luteinizing hormone (LH) in rats treated with all doses administered except with the dose of 6 mg/kg/day. PFOS also induced a raise of the follicle stimulating hormone (FSH) gene expression in the animals exposed to 0.5 and 1.0 mg of PFOS/kg/day. After PFOS exposure, hypothalamic GnRH concentration was modified, LH and testosterone release was inhibited and FSH secretion was stimulated. Moreover, PFOS induced several histopathological alterations in the hypothalamus, pituitary gland and testis. The results obtained in the present study suggest in general terms that PFOS can inhibit the physiological activity of the reproductive axis in adult male rats, which could be explained, at least in part, by the structural alterations showed in the animals exposed to this chemical: very dense chromatin, condensed ribosomes and a loss of the morphology in the hypothalamus; a degeneration of the gonadotrophic cells, as well as a loss and degeneration of the spermatozoids and a very marked edema in the testis.
Collapse
Affiliation(s)
- S López-Doval
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas S/n, 32004 Ourense, Spain
| | - R Salgado
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas S/n, 32004 Ourense, Spain
| | - N Pereiro
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas S/n, 32004 Ourense, Spain
| | - R Moyano
- Department of Pharmacology, Toxicology and Legal and Forensic Medicine, Veterinary Faculty, University of Córdoba, Córdoba 14071, Spain
| | - A Lafuente
- Laboratory of Toxicology, Sciences School, University of Vigo, Las Lagunas S/n, 32004 Ourense, Spain.
| |
Collapse
|
137
|
Lyons DJ, Broberger C. TIDAL WAVES: Network mechanisms in the neuroendocrine control of prolactin release. Front Neuroendocrinol 2014; 35:420-38. [PMID: 24561279 DOI: 10.1016/j.yfrne.2014.02.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 12/22/2013] [Accepted: 02/10/2014] [Indexed: 11/19/2022]
Abstract
Neuroendocrine tuberoinfundibular dopamine (TIDA) neurons tonically inhibit pituitary release of the hormone, prolactin. Through the powerful actions of prolactin in promoting lactation and maternal behaviour while suppressing sexual drive and fertility, TIDA neurons play a key role in reproduction. We summarize insights from recent in vitro studies into the membrane properties and network behaviour of TIDA neurons including the observations that TIDA neurons exhibit a robust oscillation that is synchronized between cells and depends on intact gap junction communication. Comparisons are made with phasic firing patterns in other neuronal populations. Modulators involved in the control of lactation - including serotonin, thyrotropin-releasing hormone and prolactin itself - have been shown to change the electrical behaviour of TIDA cells. We propose that TIDA discharge mode may play a central role in tuning the amount of dopamine delivered to the pituitary and hence circulating prolactin concentrations in different reproductive states and pathological conditions.
Collapse
Affiliation(s)
- David J Lyons
- Dept. of Neuroscience, Karolinska Institutet, Retzius v. 8, 171 77 Stockholm, Sweden
| | - Christian Broberger
- Dept. of Neuroscience, Karolinska Institutet, Retzius v. 8, 171 77 Stockholm, Sweden.
| |
Collapse
|
138
|
Yamada H, Takeda T, Koga T, Ishii Y. [Role of the critical period in sex and brain differentiation: learning from dioxin-induced disorders in next generations]. YAKUGAKU ZASSHI 2014; 134:529-35. [PMID: 24694814 DOI: 10.1248/yakushi.13-00251-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The sexual differentiation of animal fetuses and infants needs stimuli by sex steroids, which are produced in their own gonads, during a short window ('critical period') of pre- and post-natal periods. Our laboratory has conducted a series of studies focusing on the damage to next generations by dioxins. When pregnant rats are exposed to a prototype of dioxin, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD; 1 μg/kg), sexual immaturity such as defects in copulation behavior as well as growth retardation emerges in their pups. We have provided evidence that such disorders are evoked, if not all, from a transient reduction in the gonadal synthesis of sex steroids in fetuses/infants during the critical period. Our studies also revealed that TCDD initially reduces the pituitary expression of luteinizing hormone (LH) to exert the effect on steroidogenesis. Several mechanisms seem to be involved in a TCDD-induced reduction in LH expression. For example, a change in epigenetic regulation in the pituitary and impaired energy production in the hypothalamus are suggested to contribute to the above reduction. Current our study has demonstrated that a transient reduction in the pituitary-gonad axis fixes the lowered expression of hypothalamic gonadotropin-releasing hormone, resulting in defects in sexual behavior. Through these topics, we discuss the role of the critical period in differentiation and development.
Collapse
Affiliation(s)
- Hideyuki Yamada
- Graduate School of Pharmaceutical Sciences, Kyushu University
| | | | | | | |
Collapse
|
139
|
Pharmaceutical options for triggering of final oocyte maturation in ART. BIOMED RESEARCH INTERNATIONAL 2014; 2014:580171. [PMID: 25133168 PMCID: PMC4123594 DOI: 10.1155/2014/580171] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/05/2014] [Accepted: 06/28/2014] [Indexed: 11/17/2022]
Abstract
Since the pioneering days of in vitro fertilization, hCG has been the gold standard to induce final follicular maturation. We herein reviewed different pharmaceutical options for triggering of final oocyte maturation in ART. The new upcoming agent seems to be GnRHa with its potential advantages over hCG trigger. GnRHa triggering elicits a surge of gonadotropins resembling the natural midcycle surge of gonadotropins, without the prolonged action of hCG, resulting in the retrieval of more mature oocytes and a significant reduction in or elimination of OHSS as compared to hCG triggering. The induction of final follicular maturation using GnRHa represents a paradigm shift in the ovulation triggering concept in ART and, thus, a way to develop a safer IVF procedure. Kisspeptins are key central regulators of the neuroendocrine mechanisms of human reproduction, who have been shown to effectively elicit an LH surge and to induce final oocyte maturation in IVF cycles. This new trigger concept may, therefore, offer a completely new, "natural" pharmacological option for ovulation induction. Whether kisspeptins will be the future agent to trigger ovulation remains to be further explored.
Collapse
|
140
|
Uenoyama Y, Tsukamura H, Maeda KI. KNDy neuron as a gatekeeper of puberty onset. J Obstet Gynaecol Res 2014; 40:1518-26. [DOI: 10.1111/jog.12398] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/03/2014] [Indexed: 11/30/2022]
Affiliation(s)
| | - Hiroko Tsukamura
- Graduate School of Bioagricultural Sciences; Nagoya University; Nagoya
| | - Kei-ichiro Maeda
- Department of Veterinary; Medical Sciences; The University of Tokyo; Tokyo Japan
| |
Collapse
|
141
|
Neurobiology of social attachments. Neurosci Biobehav Rev 2014; 43:173-82. [DOI: 10.1016/j.neubiorev.2014.04.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 04/05/2014] [Accepted: 04/10/2014] [Indexed: 01/21/2023]
|
142
|
Handa RJ, Weiser MJ. Gonadal steroid hormones and the hypothalamo-pituitary-adrenal axis. Front Neuroendocrinol 2014; 35:197-220. [PMID: 24246855 PMCID: PMC5802971 DOI: 10.1016/j.yfrne.2013.11.001] [Citation(s) in RCA: 308] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 10/04/2013] [Accepted: 11/07/2013] [Indexed: 12/17/2022]
Abstract
The hypothalamo-pituitary-adrenal (HPA) axis represents a complex neuroendocrine feedback loop controlling the secretion of adrenal glucocorticoid hormones. Central to its function is the paraventricular nucleus of the hypothalamus (PVN) where neurons expressing corticotropin releasing factor reside. These HPA motor neurons are a primary site of integration leading to graded endocrine responses to physical and psychological stressors. An important regulatory factor that must be considered, prior to generating an appropriate response is the animal's reproductive status. Thus, PVN neurons express androgen and estrogen receptors and receive input from sites that also express these receptors. Consequently, changes in reproduction and gonadal steroid levels modulate the stress response and this underlies sex differences in HPA axis function. This review examines the make up of the HPA axis and hypothalamo-pituitary-gonadal (HPG) axis and the interactions between the two that should be considered when exploring normal and pathological responses to environmental stressors.
Collapse
Affiliation(s)
- Robert J Handa
- Department of Basic Medical Science, The University of Arizona College of Medicine, Phoenix, AZ 85004, United States.
| | - Michael J Weiser
- DSM Nutritional Products Ltd., R&D Human Nutrition and Health, Boulder, CO 80301, United States
| |
Collapse
|
143
|
Wiltbank MC, Pursley JR. The cow as an induced ovulator: Timed AI after synchronization of ovulation. Theriogenology 2014; 81:170-85. [DOI: 10.1016/j.theriogenology.2013.09.017] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Revised: 09/17/2013] [Accepted: 09/18/2013] [Indexed: 12/22/2022]
|
144
|
Jaszberenyi M, Schally AV, Block NL, Nadji M, Vidaurre I, Szalontay L, Rick FG. Inhibition of U-87 MG glioblastoma by AN-152 (AEZS-108), a targeted cytotoxic analog of luteinizing hormone-releasing hormone. Oncotarget 2013; 4:422-32. [PMID: 23518876 PMCID: PMC3717305 DOI: 10.18632/oncotarget.917] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma multiforme is the most frequent tumor of the central nervous system in adults and has a dismal clinical outcome, which necessitates the development of new therapeutic approaches. We investigated in vivo the action of the targeted cytotoxic analog of luteinizing hormone releasing hormone, AN-152 (AEZS-108) in nude mice (Ncr nu/nu strain) bearing xenotransplanted U-87 MG glioblastoma tumors. We evaluated in vitro the expression of LHRH receptors, proliferation, apoptosis and the release of oncogenic and tumor suppressor cytokines. Clinical and U-87 MG samples of glioblastoma tumors expressed LHRH receptors. Treatment of nude mice with AN-152, once a week at an intravenous dose of 413 nmol/20g, for six weeks resulted in 76 % reduction in tumor growth. AN-152 nearly completely abolished tumor progression and elicited remarkable apoptosis in vitro. Genomic (RT-PCR) and proteomic (ELISA, Western blot) studies revealed that AN-152 activated apoptosis, as reflected by the changes in p53 and its regulators and substrates, inhibited cell growth, and elicited changes in intermediary filament pattern. AN-152 similarly reestablished contact regulation as demonstrated by expression of adhesion molecules and inhibited vascularization, as reflected by the transcription of angiogenic factors. Our findings suggest that targeted cytotoxic analog AN-152 (AEZS-108) should be considered for a treatment of glioblastomas.
Collapse
|
145
|
Hodne K, Strandabø RAU, von Krogh K, Nourizadeh-Lillabadi R, Sand O, Weltzien FA, Haug TM. Electrophysiological differences between fshb- and lhb-expressing gonadotropes in primary culture. Endocrinology 2013; 154:3319-30. [PMID: 23836032 DOI: 10.1210/en.2013-1164] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Synthesis and release of FSH and LH are differentially regulated by GnRH, but the mechanisms by which this regulation is achieved are not well understood. Teleost fish are powerful models for studying this differential regulation because they have distinct pituitary cells producing either FSH or LH. By using pituitary cultures from Atlantic cod (Gadus morhua), we were able to investigate and compare the electrophysiological properties of fshb- and lhb-expressing cells, identified by single-cell quantitative PCR after recording. Both cell types fired action potentials spontaneously. The relative number of excitable cells was dependent on reproductive season but varied in opposing directions according to season in the 2 cell types. Excitable and quiescent gonadotropes displayed different ion channel repertoires. The dynamics of outward currents and GnRH-induced membrane responses differed between fshb- and lhb-expressing cells, whereas GnRH-induced cytosolic Ca²⁺ responses were similar. Expression of Ca²⁺-activated K⁺ channels also differed with cell type and showed seasonal variation when measured in whole pituitary. The differential presence of these channels corresponds to the differences observed in membrane response to GnRH. We speculate that differences in ion channel expression levels may be involved in seasonal regulation of hormone secretion as well as the differential response to GnRH in LH- and FSH-producing gonadotropes, through differences in excitability and Ca²⁺ influx.
Collapse
Affiliation(s)
- Kjetil Hodne
- Department of Basic Sciences and Aquatic Medicine, Norwegian School of Veterinary Science, N-0033 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
146
|
Nukolova NV, Oberoi HS, Zhao Y, Chekhonin VP, Kabanov AV, Bronich TK. LHRH-targeted nanogels as a delivery system for cisplatin to ovarian cancer. Mol Pharm 2013; 10:3913-21. [PMID: 23957812 DOI: 10.1021/mp4003688] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Targeted drug delivery using multifunctional polymeric nanocarriers is a modern approach for cancer therapy. Our purpose was to prepare targeted nanogels for selective delivery of chemotherapeutic agent cisplatin to luteinizing hormone-releasing hormone (LHRH) receptor overexpressing tumor in vivo. Building blocks of such delivery systems consisted of innovative soft block copolymer nanogels with ionic cores serving as a reservoir for cisplatin (loading 35%) and a synthetic analogue of LHRH conjugated to the nanogels via poly(ethylene glycol) spacer. Covalent attachment of (D-Lys6)-LHRH to nanogels was shown to be possible without loss in either the ligand binding affinity or the nanogel drug incorporation ability. LHRH-nanogel accumulation was specific to the LHRH-receptor positive A2780 ovarian cancer cells and not toward LHRH-receptor negative SKOV-3 cells. The LHRH-nanogel cisplatin formulation was more effective and less toxic than equimolar doses of free cisplatin or untargeted nanogels in the treatment of receptor-positive ovarian cancer xenografts in mice. Collectively, the study indicates that LHRH mediated nanogel-cisplatin delivery is a promising formulation strategy for therapy of tumors that express the LHRH receptor.
Collapse
Affiliation(s)
- Natalia V Nukolova
- Department of Chemistry, Lomonosov Moscow State University , Leninskie Gory, Moscow, 119992, Russia
| | | | | | | | | | | |
Collapse
|
147
|
Rick FG, Block NL, Schally AV. Agonists of luteinizing hormone-releasing hormone in prostate cancer. Expert Opin Pharmacother 2013; 14:2237-47. [PMID: 23984804 DOI: 10.1517/14656566.2013.834328] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Androgen deprivation therapy (ADT) has been the first-line standard of care for treating patients with hormone-sensitive advanced prostate cancer (PCa) for many decades. The agonists of luteinizing hormone-releasing hormone (LHRH), also called gonadotropin-releasing hormone, are still the most frequently used form of medical ADT. AREAS COVERED This article reviews the available data and most recent information concerning the use of LHRH agonists in advanced PCa. This article also reviews the discovery and development of LHRH agonists and summarizes the clinical evidence for their efficacy in PCa. EXPERT OPINION The introduction and application of agonists of LHRH has modernized and improved the treatment of advanced PCa. The life-saving benefits of LHRH agonists are well established, yet underestimated. Despite their efficacy, agonists of LHRH have several disadvantages or drawbacks including disease flare. The approach to ADT has been recently further refined with the development of the LHRH antagonist degarelix. Degarelix, a highly clinically effective third-generation LHRH antagonist, is currently available in most countries for therapy of advanced PCa. This new drug offers attractive alternatives to LHRH agonists for treatment of advanced PCa. A therapy for castration-resistant PCa based on a targeted cytotoxic analog of LHRH, AEZS-108, is also emerging.
Collapse
Affiliation(s)
- Ferenc G Rick
- Endocrine, Polypeptide, and Cancer Institute, Veterans Affairs Medical Center, South Florida Veterans Affairs Foundation for Research and Education , 1201 NW 16th St, Research (151), Room 2A103C, Miami, FL 33125 , USA +1 305 575 3477 ; +1 305 575 3126 ;
| | | | | |
Collapse
|
148
|
Ji K, Hong S, Kho Y, Choi K. Effects of bisphenol s exposure on endocrine functions and reproduction of zebrafish. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2013; 47:8793-800. [PMID: 23806087 DOI: 10.1021/es400329t] [Citation(s) in RCA: 267] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
While bisphenol S (BPS) has been frequently detected both in environment and biota, limited information is available on their effects of endocrine system. In the present study, adult zebrafish pairs were exposed to 0.5, 5, and 50 μg/L of BPS for 21 d, and the effects on reproduction, sex steroid hormones, and transcription of the genes belonging to the hypothalamic-pituitary-gonad (HPG) axis were investigated. The adverse effects on performances of F1 generation were further examined with or without subsequent exposure to BPS. Egg production and the gonadosomatic index in female fish were significantly decreased at ≥0.5 μg/L BPS. Plasma concentrations of 17β-estradiol were significantly increased in both male and female fish. In male fish, however, significant decreases of testosterone concentration were observed along with up-regulation of cyp19a and down-regulation of cyp17 and 17βhsd transcripts. Parental exposure to BPS resulted in delayed and lesser rates of hatching even when they were hatched in clean water. Continuous BPS exposure in the F1 embryos resulted in worse hatchability and increased malformation rates compared to those without BPS exposure. Our observations showed that exposure to low level BPS could affect the feedback regulatory circuits of HPG axis and impair the development of offspring.
Collapse
Affiliation(s)
- Kyunghee Ji
- School of Public Health, Seoul National University , Gwanak, Seoul 151-742, Republic of Korea
| | | | | | | |
Collapse
|
149
|
McGuire N, Ferris JK, Arckens L, Bentley GE, Soma KK. Gonadotropin releasing hormone (GnRH) and gonadotropin inhibitory hormone (GnIH) in the songbird hippocampus: regional and sex differences in adult zebra finches. Peptides 2013; 46:64-75. [PMID: 23727031 DOI: 10.1016/j.peptides.2013.05.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 05/14/2013] [Accepted: 05/15/2013] [Indexed: 11/15/2022]
Abstract
Hypothalamic gonadotropin releasing hormone (GnRH) and gonadotropin inhibitory hormone (GnIH) are vital to reproduction in all vertebrates. These neuropeptides are also present outside of the hypothalamus, but the roles of extra-hypothalamic GnRH and GnIH remain enigmatic and widely underappreciated. We used immunohistochemistry and PCR to examine whether multiple forms of GnRH (chicken GnRH-I (GnRH1), chicken GnRH-II (GnRH2) and lamprey GnRH-III (GnRH4)) and GnIH are present in the hippocampus (Hp) of adult zebra finches (Taeniopygia guttata). Using immunohistochemistry, we provide evidence that GnRH1, GnRH2 and GnRH4 are present in hippocampal cell bodies and/or fibers and that GnIH is present in hippocampal fibers only. There are regional differences in hippocampal GnRH immunoreactivity, and these vary across the different forms of GnRH. There are also sex differences in hippocampal GnRH immunoreactivity, with generally more GnRH1 and GnRH2 in the female Hp. In addition, we used PCR to examine the presence of GnRH1 mRNA and GnIH mRNA in micropunches of Hp. PCR and subsequent product sequencing demonstrated the presence of GnRH1 mRNA and the absence of GnIH mRNA in the Hp, consistent with the pattern of immunohistochemical results. To our knowledge, this is the first study in any species to systematically examine multiple forms of GnRH in the Hp or to quantify sex or regional differences in hippocampal GnRH. Moreover, this is the first demonstration of GnIH in the avian Hp. These data shed light on an important issue: the sites of action and possible functions of GnRH and GnIH outside of the HPG axis.
Collapse
Affiliation(s)
- Nicolette McGuire
- Department of Integrative Biology, Helen Wills Neuroscience Institute, University of California-Berkeley, Berkeley, CA, USA
| | | | | | | | | |
Collapse
|
150
|
Mita M. Release of Relaxin-Like Gonad-Stimulating Substance from Starfish Radial Nerves by lonomycin. Zoolog Sci 2013; 30:602-6. [DOI: 10.2108/zsj.30.602] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|