51
|
Establishment of an mKate2-Expressing Cell Line for Non-Invasive Real-Time Breast Cancer In Vivo Imaging. Mol Imaging Biol 2016; 17:811-8. [PMID: 25902968 DOI: 10.1007/s11307-015-0853-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Non-invasive real-time in vivo imaging experiments using mice as animal models have become crucial for understanding cancer development and treatment. In this study, we have developed and validated a new breast cancer cell line MDA-MB-435s that stably express a far-red fluorescence protein (mKate2) and that could serve as a highly valuable cell model for studying breast cancer detection and therapy using in vivo fluorescence imaging in nude mice. PROCEDURES The new cell line (MDA-MB-435s-mKate2) was constructed by plasmid transfection. The stability and sensitivity of mKate2, and the cell biological activities, were tested in vitro using different experimental approaches. For its potential use in tumor growth research and drug therapy in vivo, MDA-MB-435s-mKate2 was validated using the immunocompromised Balb/c nude mice tumor model. In addition, the new cell line has been characterized as a luteinizing hormone-releasing hormone receptor (LHRHR) positive cell line. RESULTS Firstly, MDA-MB-435s-mKate2 has shown a stable chromosomal integration of the amplified mKate2 gene and good fluorescence sensitivity for detection using a fluorescence reflectance imaging (FRI) device. Compared to its parental cell line, no significant difference in cell migration, proliferation, and clone formation was observed in vitro. Secondly, using the quantification of tumor-fluorescence surface area in live animals, we were able to monitor and detect the tumor progress or tumor inhibition rate (by Paclitaxel treatment) non-invasively and in real-time. Furthermore, MDA-MB-435s-mKate2 has been positively tested for LHRHR; these findings open the possibility to use this cell line for future studies of breast cancer therapy based on LHRH analogs in vivo. CONCLUSION In the present research, we have successfully built the MDA-MB-435s-mKate2 cell line that can be used as a suitable cell model for breast cancer therapy and anti-cancer drug evaluation by non-invasive fluorescence imaging in mice.
Collapse
|
52
|
Aguilar-Rojas A, Pérez-Solis MA, Maya-Núñez G. The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review). Int J Oncol 2016; 48:861-8. [PMID: 26783137 DOI: 10.3892/ijo.2016.3346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 11/06/2022] Open
Abstract
Recently, an increasing amount of evidence indicates that human gonadotropin-releasing hormone (hGnRH) and its receptor (hGnRHR) are important regulatory components not only to the reproduction process but also in the regulation of some cancer cell functions such as cell proliferation, in both hormone-dependent and -independent types of tumors. The hGnRHR is a naturally misfolded protein that is retained mostly in the endoplasmic reticulum; however, this mechanism can be overcome by treatment with several pharmacoperones, therefore, increasing the amount of receptors in the cell membrane. In addition, several reports indicate that the expression level of hGnRHR in tumor cells is even lower than in pituitary or gonadotrope cells. The signal transduction pathways activated by hGnRH in both gonadotrope and different cancer cell types are described in the present review. We also discuss how the rescue of misfolded receptors in tumor cells could be a promising strategy for cancer therapy.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Research Unit in Reproductive Medicine, Health Research Council, Hospital de Gineco-Obstetricia 'Luis Castelazo Ayala', Instituto Mexicano del Seguro Social, Mexico 01090, D.F., Mexico
| | - Marco Allan Pérez-Solis
- Research Unit in Reproductive Medicine, Health Research Council, Hospital de Gineco-Obstetricia 'Luis Castelazo Ayala', Instituto Mexicano del Seguro Social, Mexico 01090, D.F., Mexico
| | - Guadalupe Maya-Núñez
- Research Unit in Reproductive Medicine, Health Research Council, Hospital de Gineco-Obstetricia 'Luis Castelazo Ayala', Instituto Mexicano del Seguro Social, Mexico 01090, D.F., Mexico
| |
Collapse
|
53
|
Guzmán-Soto I, Salinas E, Quintanar JL. Leuprolide Acetate Inhibits Spinal Cord Inflammatory Response in Experimental Autoimmune Encephalomyelitis by Suppressing NF-κB Activation. Neuroimmunomodulation 2016; 23:33-40. [PMID: 26445405 DOI: 10.1159/000438927] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/17/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Recent findings have shown that gonadotropin-releasing hormone (GnRH) administration in an animal model of multiple sclerosis (experimental autoimmune encephalomyelitis, EAE) improves clinical signs of locomotion. The present study was designed to determine whether the administration of the synthetic analog of GnRH, leuprolide acetate (LA) - besides its effects on clinical signs of locomotion - also has an effect on the activation/expression levels of molecular markers of EAE, namely transcription nuclear factor (NF)-κB and the proinflammatory cytokines IL-1β, IL-17A, IL-23 and TNF-α. METHODS EAE spinal cords were collected from control and LA-administered rats. Lumbar sections were processed at four different time points during the course of the disease to analyze NF-κB activation by chemiluminescent Western blot, and during the EAE recovery phase to evaluate proinflammatory cytokine levels by quantitative real-time PCR. RESULTS It was found that LA administration to EAE rats promoted a significant reduction of NF-κB activation during the course of the disease and also decreased the mRNA expression levels of the proinflammatory cytokines IL-1β, IL-17A and TNF-α in the EAE recovery phase; both effects are consistent with the decrease in the severity of clinical signs of locomotion induced by the treatment. CONCLUSION LA causes a reduction in the severity of locomotor activity, as well as in the activation of NF-κB and the number of proinflammatory markers in rats with EAE. These results suggest the use of this agonist as a potential therapeutic approach for multiple sclerosis.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Bx00E1;sicas, Universidad Autx00F3;noma de Aguascalientes, Aguascalientes, Mexico
| | | | | |
Collapse
|
54
|
Maggi R, Cariboni AM, Marelli MM, Moretti RM, Andrè V, Marzagalli M, Limonta P. GnRH and GnRH receptors in the pathophysiology of the human female reproductive system. Hum Reprod Update 2015; 22:358-81. [PMID: 26715597 DOI: 10.1093/humupd/dmv059] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 12/03/2015] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Human reproduction depends on an intact hypothalamic-pituitary-gonadal (HPG) axis. Hypothalamic gonadotrophin-releasing hormone (GnRH) has been recognized, since its identification in 1971, as the central regulator of the production and release of the pituitary gonadotrophins that, in turn, regulate the gonadal functions and the production of sex steroids. The characteristic peculiar development, distribution and episodic activity of GnRH-producing neurons have solicited an interdisciplinary interest on the etiopathogenesis of several reproductive diseases. The more recent identification of a GnRH/GnRH receptor (GnRHR) system in both the human endometrium and ovary has widened the spectrum of action of the peptide and of its analogues beyond its hypothalamic function. METHODS An analysis of research and review articles published in international journals until June 2015 has been carried out to comprehensively summarize both the well established and the most recent knowledge on the physiopathology of the GnRH system in the central and peripheral control of female reproductive functions and diseases. RESULTS This review focuses on the role of GnRH neurons in the control of the reproductive axis. New knowledge is accumulating on the genetic programme that drives GnRH neuron development to ameliorate the diagnosis and treatment of GnRH deficiency and consequent delayed or absent puberty. Moreover, a better understanding of the mechanisms controlling the episodic release of GnRH during the onset of puberty and the ovulatory cycle has enabled the pharmacological use of GnRH itself or its synthetic analogues (agonists and antagonists) to either stimulate or to block the gonadotrophin secretion and modulate the functions of the reproductive axis in several reproductive diseases and in assisted reproduction technology. Several inputs from other neuronal populations, as well as metabolic, somatic and age-related signals, may greatly affect the functions of the GnRH pulse generator during the female lifespan; their modulation may offer new possible strategies for diagnostic and therapeutic interventions. A GnRH/GnRHR system is also expressed in female reproductive tissues (e.g. endometrium and ovary), both in normal and pathological conditions. The expression of this system in the human endometrium and ovary supports its physiological regulatory role in the processes of trophoblast invasion of the maternal endometrium and embryo implantation as well as of follicular development and corpus luteum functions. The GnRH/GnRHR system that is expressed in diseased tissues of the female reproductive tract (both benign and malignant) is at present considered an effective molecular target for the development of novel therapeutic approaches for these pathologies. GnRH agonists are also considered as a promising therapeutic approach to counteract ovarian failure in young female patients undergoing chemotherapy. CONCLUSIONS Increasing knowledge about the regulation of GnRH pulsatile release, as well as the therapeutic use of its analogues, offers interesting new perspectives in the diagnosis, treatment and outcome of female reproductive disorders, including tumoral and iatrogenic diseases.
Collapse
Affiliation(s)
- Roberto Maggi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Anna Maria Cariboni
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Roberta Manuela Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Valentina Andrè
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via G. Balzaretti, 9, 20133 Milano, Italy
| |
Collapse
|
55
|
Cortés-Campos C, Letelier J, Ceriani R, Whitlock KE. Zebrafish adult-derived hypothalamic neurospheres generate gonadotropin-releasing hormone (GnRH) neurons. Biol Open 2015. [PMID: 26209533 PMCID: PMC4582115 DOI: 10.1242/bio.010447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is a hypothalamic decapeptide essential for fertility in vertebrates. Human male patients lacking GnRH and treated with hormone therapy can remain fertile after cessation of treatment suggesting that new GnRH neurons can be generated during adult life. We used zebrafish to investigate the neurogenic potential of the adult hypothalamus. Previously we have characterized the development of GnRH cells in the zebrafish linking genetic pathways to the differentiation of neuromodulatory and endocrine GnRH cells in specific regions of the brain. Here, we developed a new method to obtain neural progenitors from the adult hypothalamus in vitro. Using this system, we show that neurospheres derived from the adult hypothalamus can be maintained in culture and subsequently differentiate glia and neurons. Importantly, the adult derived progenitors differentiate into neurons containing GnRH and the number of cells is increased through exposure to either testosterone or GnRH, hormones used in therapeutic treatment in humans. Finally, we show in vivo that a neurogenic niche in the hypothalamus contains GnRH positive neurons. Thus, we demonstrated for the first time that neurospheres can be derived from the hypothalamus of the adult zebrafish and that these neural progenitors are capable of producing GnRH containing neurons.
Collapse
Affiliation(s)
- Christian Cortés-Campos
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile Whitehead Institute for Biomedical Research (WIBR), 9 Cambridge Center, Cambridge, MA 02142, USA
| | - Joaquín Letelier
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Carretera de Utera km 1, Sevilla 41013, España
| | - Ricardo Ceriani
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile
| | - Kathleen E Whitlock
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Facultad de Ciencias, Universidad de Valparaíso, Pasaje Harrington 269, Valparaíso 2340000, Chile
| |
Collapse
|
56
|
Teng LH, Ahmad M, Ng WTW, Sabaratnam S, Rasan MI, Parhar I, Khoo ASB. Gonadotropin‑releasing hormone inhibits the proliferation and motility of nasopharyngeal carcinoma cells. Mol Med Rep 2015; 12:4909-16. [PMID: 26151677 PMCID: PMC4581822 DOI: 10.3892/mmr.2015.4043] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 06/05/2015] [Indexed: 11/07/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH), or its analogues have been demonstrated to exhibit anti-proliferative effects on tumour cells in ovarian, endometrial and breast cancer through GnRH-receptors (GnRH-R). However, the role of GnRH in nasopharyngeal carcinoma (NPC) remains to be elucidated. In order to investigate the effects of GnRH in NPC, the present study examined the expression of the GnRH-R transcript in NPC and investigated the phenotypic changes in HK1 cells, a recurrent NPC-derived cell line, upon receiving GnRH treatment. Firstly, the GnRH-R transcript was demonstrated in the NPC cell lines and four snap frozen biopsies using reverse transcription-quantitative polymerase chain reaction. In addition, immunohistochemistry revealed the expression of GnRH-R in two of the eight (25%) NPC specimens. Treatment with GnRH induced a rapid increase in intracellular ionised calcium concentration in the NPC cells. GnRH and its agonists, triptorelin and leuprolide, exerted anti-proliferative effects on the NPC cells, as determined using an MTS assay. GnRH did not induce any cell cycle arrest in the HK1 cells under the conditions assessed in the present study. Time-lapse imaging demonstrated a reduction in cell motility in the GnRH-treated cells. In conclusion, GnRH, or its analogues may have antitumour effects on NPC cells. The consequences of alterations in the levels of GnRH on the progression of NPC require further examination.
Collapse
Affiliation(s)
- Loong Hung Teng
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia
| | - Munirah Ahmad
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia
| | - Wayne Tiong Weng Ng
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia
| | - Subathra Sabaratnam
- Department of Pathology, Sultanah Aminah Hospital, Jeffrey Cheah's School of Medicine and Health Sciences, Monash University, Johor Bahru, Johor 80100, Malaysia
| | - Maria Ithaya Rasan
- Department of Otorhinolaryngology, Hospital Tengku Ampuan Rahimah, Klang, Selangor 41200, Malaysia
| | - Ishwar Parhar
- Brain Research Institute, School of Medicine and Health Sciences, Monash University Sunway Campus, Petaling Jaya, Selangor 47500, Malaysia
| | - Alan Soo Beng Khoo
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, Kuala Lumpur 50588, Malaysia
| |
Collapse
|
57
|
Lu C, Huang T, Chen W, Lu H. GnRH participates in the self-renewal of A549-derived lung cancer stem-like cells through upregulation of the JNK signaling pathway. Oncol Rep 2015; 34:244-50. [PMID: 25955300 DOI: 10.3892/or.2015.3956] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 02/03/2015] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality in humans. Exploration of the mechanisms underlying the self-renewal and stemness maintenance of cancer stem-like cells (CSLCs) will open new avenues in lung cancer diagnosis and therapy. Here, we isolated and identified a subpopulation of lung cancer stem-like cells (LCSLCs) from non-small cell lung carcinoma (NSCLC) A549 cells with features including self-renewal capacity in vitro, elevated tumorigenic activity in vivo, and high expression of stemness markers CD44, CD133, aldehyde dehydrogenase 1 (ALDH1) and Sox2, using a serum-free suspension sphere-forming culture method. We then found a higher expression level of gonadotropin-releasing hormone (GnRH) in the LCSLCs using a microarray assay, suggesting that GnRH may play a role in the self-renewal capacity and stemness maintenance in lung cancer cells. In addition, the suppression of GnRH capacity negatively regulated self-renewal and stemness maintenance in the LCSLCs. Overexpression of GnRH promoted stemness properties of A549-derived LCSLCs, indicating that GnRH expression is essential for the self-renewal and stemness maintenance in LCSLCs. Moreover, further investigations demonstrated that the promotion of GnRH functions of self-renewal and stemness maintenance in LCSLCs was associated with the JNK signaling pathway. Therefore, our results showed that GnRH participates in the self-renewal capacity and stemness maintenance of LCSLCs by upregulating the JNK signaling pathway, and GnRH may be useful as an alternative LCSLC therapy.
Collapse
Affiliation(s)
- Chi Lu
- Department of Oncology, The Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Ting Huang
- Department of Oncology, The Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Weiqun Chen
- Cancer Research Institute of Wuhan, Wuhan, Hubei 430014, P.R. China
| | - Hongda Lu
- Department of Oncology, The Central Hospital of Wuhan, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
58
|
Szabó I, Bősze S, Orbán E, Sipos É, Halmos G, Kovács M, Mező G. Comparative in vitro biological evaluation of daunorubicin containing GnRH-I and GnRH-II conjugates developed for tumor targeting. J Pept Sci 2015; 21:426-35. [PMID: 25908176 DOI: 10.1002/psc.2775] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/27/2015] [Accepted: 03/04/2015] [Indexed: 12/22/2022]
Abstract
Hormone based drug targeting is a promising tool for selective tumor therapy. In this study, synthesis and systematic comparative biological evaluation of novel drug containing analogs of gonadotropin-releasing hormone GnRH-I and GnRH-II is reported demonstrating their suitability for tumor targeting. The cytotoxic conjugates were prepared by the attachment of the chemotherapeutical agent daunorubicin (Dau) to GnRH analogs directly or through an enzyme-labile spacer with oxime linkage. All conjugates were found to be proteolytically stable under circumstances applied in biological assays. Both GnRH-I and GnRH-II were able to bind similarly to high-affinity GnRH-I receptors on human pituitary and human prostate cancer cells. The in vitro long-term cytotoxic effect of the conjugates was comparable with that of the free drug in human breast and colon cancer cell lines. Furthermore, a concentration-dependent cellular uptake profile was observed. The in vitro apoptotic effect of the compounds was evaluated by flow cytometry analysis using annexin-V. Our results show that both the GnRH-I and the GnRH-II based analogs might be applied for targeted tumor therapy.
Collapse
Affiliation(s)
- Ildikó Szabó
- MTA-ELTE Research Group of Peptide Chemistry, Hungarian Academy of Sciences, Eötvös L. University, 1117, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
59
|
Sakai M, Martinez-Arguelles DB, Patterson NH, Chaurand P, Papadopoulos V. In search of the molecular mechanisms mediating the inhibitory effect of the GnRH antagonist degarelix on human prostate cell growth. PLoS One 2015; 10:e0120670. [PMID: 25811175 PMCID: PMC4374753 DOI: 10.1371/journal.pone.0120670] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 02/05/2015] [Indexed: 12/13/2022] Open
Abstract
Degarelix is a gonadrotropin-releasing hormone (GnRH) receptor (GnRHR) antagonist used in patients with prostate cancer who need androgen deprivation therapy. GnRHRs have been found in extra-pituitary tissues, including prostate, which may be affected by the GnRH and GnRH analogues used in therapy. The direct effect of degarelix on human prostate cell growth was evaluated. Normal prostate myofibroblast WPMY-1 and epithelial WPE1-NA22 cells, benign prostatic hyperplasia (BPH)-1 cells, androgen-independent PC-3 and androgen-dependent LNCaP prostate cancer cells, as well as VCaP cells derived from a patient with castration-resistant prostate cancer were used. Discriminatory protein and lipid fingerprints of normal, hyperplastic, and cancer cells were generated by matrix-assisted laser desorption/ionization (MALDI) mass spectrometry (MS). The investigated cell lines express GNRHR1 and GNRHR2 and their endogenous ligands. Degarelix treatment reduced cell viability in all prostate cell lines tested, with the exception of the PC-3 cells; this can be attributed to increased apoptosis, as indicated by increased caspase 3/7, 8 and 9 levels. WPE1-NA22, BPH-1, LNCaP, and VCaP cell viability was not affected by treatment with the GnRH agonists leuprolide and goserelin. Using MALDI MS, we detected changes in m/z signals that were robust enough to create a complete discriminatory profile induced by degarelix. Transcriptomic analysis of BPH-1 cells provided a global map of genes affected by degarelix and indicated that the biological processes affected were related to cell growth, G-coupled receptors, the mitogen-activated protein kinase (MAPK) pathway, angiogenesis and cell adhesion. Taken together, these data demonstrate that (i) the GnRH antagonist degarelix exerts a direct effect on prostate cell growth through apoptosis; (ii) MALDI MS analysis provided a basis to fingerprint degarelix-treated prostate cells; and (iii) the clusters of genes affected by degarelix suggest that this compound, in addition to its known use in the treatment of prostate cancer, may be efficacious in BPH.
Collapse
Affiliation(s)
- Monica Sakai
- The Research Institute of the McGill University Health Center, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | - Daniel B. Martinez-Arguelles
- The Research Institute of the McGill University Health Center, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
| | | | - Pierre Chaurand
- Department of Chemistry, University of Montreal, Montréal, Québec, Canada
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Center, Montréal, Québec, Canada
- Department of Medicine, McGill University, Montréal, Québec, Canada
- Departments of Biochemistry, McGill University, Montréal, Québec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
60
|
Wu X, Yan M, Wan X, Lian S, Li A. Cloning and tissue expression profiling of theGnRHRgene of the Muscovy duck (Cairina moschata): polymorphism and association with egg-laying performance. Br Poult Sci 2015; 56:164-74. [DOI: 10.1080/00071668.2014.997674] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
61
|
Calderón-Vallejo D, Quintanar-Stephano A, Hernández-Jasso I, Jiménez-Hernández V, Ruiz-Ornelas J, Jiménez I, Quintanar JL. Functional and structural recovery of the injured spinal cord in rats treated with gonadotropin-releasing hormone. Neurochem Res 2015; 40:455-62. [PMID: 25618391 DOI: 10.1007/s11064-014-1486-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 10/24/2014] [Accepted: 11/20/2014] [Indexed: 12/19/2022]
Abstract
Several studies have shown that gonadotropin-releasing hormone (GnRH) have extra-pituitary roles, including neurotrophic effects. This study was to evaluate the effects of GnRH treatment on the spinal cord injury (SCI) of rats. Ovariectomized rats were divided into: sham SCI surgery (Sham), SCI treated with saline solution (SCI + SS), and SCI treated with GnRH (SCI + GnRH). The SCI was induced by compression. One day after the lesion, SCI + GnRH group was injected with GnRH (60 µg/kg/twice/day; i.m.) for 15 days and the other groups with saline solution. To kinematic gait analysis, length and velocity of the stride were measured. In spinal cord, axonal morphometry and spared white and gray matter were analyzed by histochemistry. Protein expression of spinophilin was evaluated by western blot. The results showed that, 5 weeks after the injury, the group of animals treated with GnRH, significantly increased the length and velocity of the stride compared to SCI + SS group and they were similar to Sham group. In spinal cord, GnRH treatment increased the number and caliber of nerve axons and in the case of white matter, spared tissue was significantly higher than those animals treated with saline solution. The expression of spinophilin in spinal cord of SCI + GnRH group was slightly increased with respect to those not treated. In conclusion, GnRH treatment improves recovery of gait and decreases histopathological damage in the injured spinal cord of rat. These findings suggest that GnRH acts as a neurotrophic factor and can be used as a potential therapeutic agent for treatment of SCI.
Collapse
Affiliation(s)
- Denisse Calderón-Vallejo
- Laboratory of Neurophysiology, Depto. de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, Col. Ciudad Universitaria, C.P. 20131, Aguascalientes, AGS, Mexico
| | | | | | | | | | | | | |
Collapse
|
62
|
Oszkiel H, Wilczak J, Jank M. Biologically active substances-enriched diet regulates gonadotrope cell activation pathway in liver of adult and old rats. GENES AND NUTRITION 2014; 9:427. [PMID: 25156242 PMCID: PMC4172640 DOI: 10.1007/s12263-014-0427-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 08/11/2014] [Indexed: 01/23/2023]
Abstract
According to the Hippocrates’ theorem “Let food be your medicine and medicine be your food”, dietary interventions may induce changes in the metabolic and inflammatory state by modulating the expression of important genes involved in the chronic disorders. The aim of the present study was to evaluate the influence of long-term (14 months) use of biologically active substances-enriched diet (BASE-diet) on transcriptomic profile of rats’ liver. The experiment was conducted on 36 Sprague–Dawley rats divided into two experimental groups (fed with control or BASE-diet, both n = 18). Control diet was a semi-synthetic diet formulated according to the nutritional requirements for laboratory animals. The BASE-diet was enriched with a mixture of polyphenolic compounds, β-carotene, probiotics, and n-3 and n-6 polyunsaturated fatty acids. In total, n = 3,017 differentially expressed (DE) genes were identified, including n = 218 DE genes between control and BASE groups after 3 months of feeding and n = 1,262 after 14 months. BASE-diet influenced the expression of genes involved particularly in the gonadotrope cell activation pathway and guanylate cyclase pathway, as well as in mast cell activation, gap junction regulation, melanogenesis and apoptosis. Especially genes involved in regulation of GnRH were strongly affected by BASE-diet. This effect was stronger with the age of animals and the length of diet use. It may suggest a link between the diet, reproductive system function and aging.
Collapse
Affiliation(s)
- Hanna Oszkiel
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Nowoursynowska 159 Str., 02-776, Warsaw, Poland
| | | | | |
Collapse
|
63
|
Karampelas T, Argyros O, Sayyad N, Spyridaki K, Pappas C, Morgan K, Kolios G, Millar RP, Liapakis G, Tzakos AG, Fokas D, Tamvakopoulos C. GnRH-Gemcitabine Conjugates for the Treatment of Androgen-Independent Prostate Cancer: Pharmacokinetic Enhancements Combined with Targeted Drug Delivery. Bioconjug Chem 2014; 25:813-23. [DOI: 10.1021/bc500081g] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Theodoros Karampelas
- Division
of Pharmacology-Pharmacotechnology, Biomedical Research Foundation
of the Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| | - Orestis Argyros
- Division
of Pharmacology-Pharmacotechnology, Biomedical Research Foundation
of the Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| | | | - Katerina Spyridaki
- Department
of Pharmacology, Faculty of Medicine, University of Crete, Heraklion, Crete, GR-71003, Greece
| | | | - Kevin Morgan
- Respiratory
Medicine, Castle Hill Hospital, Hull York Medical School, University of Hull, Hull, Yorkshire HU6 7RX, United Kingdom
| | - George Kolios
- Laboratory
of Pharmacology, School of Medicine, Democritus University of Thrace, Alexandroupolis, GR-68100, Greece
| | - Robert P Millar
- Mammal
Research Institute, Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa
- UCT/MRC
Receptor Biology Group, Institute for Infectious Diseases and Molecular
Medicine, University of Cape Town, Rondebosch 7701, South Africa
- Centre
for Integrative Medicine, University of Edinburgh, Edinburgh EH8 9YL, Scotland
| | - George Liapakis
- Department
of Pharmacology, Faculty of Medicine, University of Crete, Heraklion, Crete, GR-71003, Greece
| | | | | | - Constantin Tamvakopoulos
- Division
of Pharmacology-Pharmacotechnology, Biomedical Research Foundation
of the Academy of Athens, Soranou Ephessiou Street 4, Athens GR-11527, Greece
| |
Collapse
|
64
|
Ellsworth DL, Croft DT, Weyandt J, Sturtz LA, Blackburn HL, Burke A, Haberkorn MJ, McDyer FA, Jellema GL, van Laar R, Mamula KA, Chen Y, Vernalis MN. Intensive Cardiovascular Risk Reduction Induces Sustainable Changes in Expression of Genes and Pathways Important to Vascular Function. ACTA ACUST UNITED AC 2014; 7:151-60. [DOI: 10.1161/circgenetics.113.000121] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background—
Healthy lifestyle changes are thought to mediate cardiovascular disease risk through pathways affecting endothelial function and progression of atherosclerosis; however, the extent, persistence, and clinical significance of molecular change during lifestyle modification are not well known. We examined the effect of a rigorous cardiovascular disease risk reduction program on peripheral blood gene expression profiles in 63 participants and 63 matched controls to characterize molecular responses and identify regulatory pathways important to cardiovascular health.
Methods and Results—
Dramatic changes in dietary fat intake (−61%;
P
<0.001 versus controls) and physical fitness (+34%;
P
<0.001) led to significant improvements in cardiovascular disease risk factors. Analysis of variance with false discovery rate correction for multiple testing (
P
<0.05) identified 26 genes after 12 weeks and 143 genes after 52 weeks that were differentially expressed from baseline in participants. Controls showed little change in cardiovascular disease risk factors or gene expression. Quantitative reverse transcription polymerase chain reaction validated differential expression for selected transcripts. Lifestyle modification effectively reduced expression of proinflammatory genes associated with neutrophil activation and molecular pathways important to vascular function, including cytokine production, carbohydrate metabolism, and steroid hormones. Prescription medications did not significantly affect changes in gene expression.
Conclusions—
Successful and sustained modulation of gene expression through lifestyle changes may have beneficial effects on the vascular system not apparent from traditional risk factors. Healthy lifestyles may restore homeostasis to the leukocyte transcriptome by downregulating lactoferrin and other genes important in the pathogenesis of atherosclerosis.
Clinical Trial Registration—
URL:
www.clinicaltrials.gov
. Unique identifier: NCT01805492
Collapse
Affiliation(s)
- Darrell L. Ellsworth
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Daniel T. Croft
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Jamie Weyandt
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Lori A. Sturtz
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Heather L. Blackburn
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Amy Burke
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Mary Jane Haberkorn
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Fionnuala A. McDyer
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Gera L. Jellema
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Ryan van Laar
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Kimberly A. Mamula
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Yaqin Chen
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| | - Marina N. Vernalis
- From the Integrative Cardiac Health Program (D.L.E., D.T.C., J.W., L.A.S., H.L.B., K.A.M.) and Clinical Breast Care Project (Y.C.), Windber Research Institute, Windber Medical Center, Windber, PA (A.B., M.J.H.); Almac Diagnostics, Craigavon, UK (F.A.M., G.L.J.); ChipDX, New York, NY (R.v.L.); and Integrative Cardiac Health Program, Walter Reed National Military Medical Center, Bethesda, MD (M.N.V.)
| |
Collapse
|
65
|
Lainas GT, Kolibianakis EM, Sfontouris IA, Zorzovilis IZ, Petsas GK, Lainas TG, Tarlatzis BC. Serum vascular endothelial growth factor levels following luteal gonadotrophin-releasing hormone antagonist administration in women with severe early ovarian hyperstimulation syndrome. BJOG 2014; 121:848-55. [DOI: 10.1111/1471-0528.12572] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2013] [Indexed: 11/29/2022]
Affiliation(s)
- GT Lainas
- Eugonia Unit of Assisted Reproduction; Athens Greece
| | - EM Kolibianakis
- Unit for Human Reproduction; 1st Department of Obstetrics and Gynaecology; Papageorgiou General Hospital; Medical School; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - IA Sfontouris
- Eugonia Unit of Assisted Reproduction; Athens Greece
| | - IZ Zorzovilis
- Eugonia Unit of Assisted Reproduction; Athens Greece
| | - GK Petsas
- Eugonia Unit of Assisted Reproduction; Athens Greece
| | - TG Lainas
- Eugonia Unit of Assisted Reproduction; Athens Greece
| | - BC Tarlatzis
- Unit for Human Reproduction; 1st Department of Obstetrics and Gynaecology; Papageorgiou General Hospital; Medical School; Aristotle University of Thessaloniki; Thessaloniki Greece
| |
Collapse
|
66
|
Zhang J, Chtcheglova LA, Zhu R, Hinterdorfer P, Zhang B, Tang J. Nanoscale Organization of Human GnRH-R on Human Bladder Cancer Cells. Anal Chem 2014; 86:2458-64. [DOI: 10.1021/ac403304g] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Jing Zhang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Science, Changchun 130022, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Lilia A. Chtcheglova
- Center
for Advanced
Bioanalysis GmbH (CBL), Gruberstrasse
40-42, Linz 4020, Austria
| | - Rong Zhu
- Institute
for Biophysics, Kepler Johannes University of Linz, Gruberstrasse
40-42, Linz 4020, Austria
| | - Peter Hinterdorfer
- Center
for Advanced
Bioanalysis GmbH (CBL), Gruberstrasse
40-42, Linz 4020, Austria
- Institute
for Biophysics, Kepler Johannes University of Linz, Gruberstrasse
40-42, Linz 4020, Austria
| | - Bailin Zhang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Science, Changchun 130022, P.R. China
| | - Jilin Tang
- State
Key Laboratory of Electroanalytical Chemistry, Changchun Institute
of Applied Chemistry, Chinese Academy of Science, Changchun 130022, P.R. China
| |
Collapse
|
67
|
Perrett RM, McArdle CA. Molecular mechanisms of gonadotropin-releasing hormone signaling: integrating cyclic nucleotides into the network. Front Endocrinol (Lausanne) 2013; 4:180. [PMID: 24312080 PMCID: PMC3834291 DOI: 10.3389/fendo.2013.00180] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 11/06/2013] [Indexed: 01/21/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is the primary regulator of mammalian reproductive function in both males and females. It acts via G-protein coupled receptors on gonadotropes to stimulate synthesis and secretion of the gonadotropin hormones luteinizing hormone and follicle-stimulating hormone. These receptors couple primarily via G-proteins of the Gq/ll family, driving activation of phospholipases C and mediating GnRH effects on gonadotropin synthesis and secretion. There is also good evidence that GnRH causes activation of other heterotrimeric G-proteins (Gs and Gi) with consequent effects on cyclic AMP production, as well as for effects on the soluble and particulate guanylyl cyclases that generate cGMP. Here we provide an overview of these pathways. We emphasize mechanisms underpinning pulsatile hormone signaling and the possible interplay of GnRH and autocrine or paracrine regulatory mechanisms in control of cyclic nucleotide signaling.
Collapse
Affiliation(s)
- Rebecca M. Perrett
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Craig A. McArdle
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
- *Correspondence: Craig A. McArdle, Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, 1 Whitson Street, Bristol BS1 3NY, UK e-mail:
| |
Collapse
|
68
|
Savulescu D, Feng J, Ping YS, Mai O, Boehm U, He B, O'Malley BW, Melamed P. Gonadotropin-releasing hormone-regulated prohibitin mediates apoptosis of the gonadotrope cells. Mol Endocrinol 2013; 27:1856-70. [PMID: 24085822 DOI: 10.1210/me.2013-1210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH regulates circulating levels of the gonadotropins but has also been implicated in establishing the gonadotrope cell population. Consistent with this, GnRH induces proliferation of partially differentiated gonadotropes, while reducing the numbers of fully differentiated cells. We have previously reported that the proapoptotic protein, prohibitin (PHB) is expressed more abundantly in gonadotrope-derived LβT2 cells than in partially differentiated αT3-1 gonadotrope precursor cells, suggesting a possible role for PHB in GnRH-induced apoptosis. We show here that PHB is required for GnRH-induced apoptosis in mature gonadotropes. PHB expression and activity are regulated by GnRH: its transcription is via c-Jun NH2-terminal kinase, whereas its nuclear export follows activation of ERK. Moreover, PHB levels are down-regulated by microRNA27, which is expressed at lower levels in mature gonadotropes, possibly explaining the switch to an apoptotic response with development. PHB is required for mitochondrial import of the proapoptotic BAX, whose expression is also induced by GnRH-activated c-Jun NH2-terminal kinase, as is expression of the BH3-only protein, HRK, and this too plays a role in GnRH-induced apoptosis. Finally, we show that gonadotrope-specific PHB-knockout mice display reproductive abnormalities, including a larger gonadotrope population, increased LH levels, reduced fertility, and altered gonad development. We thus demonstrate a role for PHB in GnRH-induced cell death in mature gonadotropes, which is crucial for the normal development and function of the reproductive axis.
Collapse
Affiliation(s)
- Dana Savulescu
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000 Israel.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Torrealday S, Lalioti MD, Guzeloglu-Kayisli O, Seli E. Characterization of the gonadotropin releasing hormone receptor (GnRHR) expression and activity in the female mouse ovary. Endocrinology 2013; 154:3877-87. [PMID: 23913446 PMCID: PMC3776864 DOI: 10.1210/en.2013-1341] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH agonists (GnRHa) are increasingly used for fertility preservation in women undergoing gonadotoxic chemotherapy. However, the protective mechanisms of action for these compounds have not yet been elucidated. In this study, we aimed to determine whether GnRHa have a direct effect on ovarian granulosa cells. GnRH receptor (GnRHR) expression was determined in mouse somatic and gonadal tissues including granulosa/cumulus cells and oocytes using quantitative RT-PCR and immunohistochemistry. Granulosa cells were isolated from mouse ovaries primed with pregnant mare serum gonadotropin. Response to GnRHa in cultured granulosa cells was assessed by determining the increase of intracellular cAMP and by assessing phosphorylation of downstream mediators of GnRH signaling: ERK and p38. To measure intracellular cAMP in our system, the cells were transfected with a cAMP-responsive luciferase reporter plasmid and stimulated with GnRHa. For all experiments, pituitary tissue and/or the αT3-1 mouse pituitary cell line were used as controls. GnRHR mRNA and protein were detected in mouse ovaries, granulosa/cumulus cells, and oocytes. After GnRHa stimulation at various time intervals, we were unable to detect a cAMP increase or activation of the ERK or p38 signaling pathway in cultured primary mouse granulosa cells, whereas activation was detected in the control αT3-1 mouse pituitary cells. In this study, we have not detected activation of the canonical GnRH signaling pathways in mouse ovarian somatic cells. Our findings suggest that the mechanism of action of GnRHa in the ovary is either below the detection level of our experimental design or is different from that in the pituitary.
Collapse
Affiliation(s)
- Saioa Torrealday
- MD, Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 310 Cedar Street, LSOG 304B, New Haven, Connecticut 06520-8063.
| | | | | | | |
Collapse
|
70
|
Limonta P, Manea M. Gonadotropin-releasing hormone receptors as molecular therapeutic targets in prostate cancer: Current options and emerging strategies. Cancer Treat Rev 2013; 39:647-63. [DOI: 10.1016/j.ctrv.2012.12.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 12/03/2012] [Indexed: 12/28/2022]
|
71
|
Quintanar JL, Guzmán-Soto I. Hypothalamic neurohormones and immune responses. Front Integr Neurosci 2013; 7:56. [PMID: 23964208 PMCID: PMC3741963 DOI: 10.3389/fnint.2013.00056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/16/2013] [Indexed: 01/19/2023] Open
Abstract
The aim of this review is to provide a comprehensive examination of the current literature describing the neural-immune interactions, with emphasis on the most recent findings of the effects of neurohormones on immune system. Particularly, the role of hypothalamic hormones such as Thyrotropin-releasing hormone (TRH), Corticotropin-releasing hormone (CRH) and Gonadotropin-releasing hormone (GnRH). In the past few years, interest has been raised in extrapituitary actions of these neurohormones due to their receptors have been found in many non-pituitary tissues. Also, the receptors are present in immune cells, suggesting an autocrine or paracrine role within the immune system. In general, these neurohormones have been reported to exert immunomodulatory effects on cell proliferation, immune mediators release and cell function. The implications of these findings in understanding the network of hypothalamic neuropeptides and immune system are discussed.
Collapse
Affiliation(s)
- J Luis Quintanar
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes Aguascalientes, México
| | | |
Collapse
|
72
|
Pregnancy and neonatal outcomes following luteal GnRH antagonist administration in patients with severe early OHSS. Hum Reprod 2013; 28:1929-42. [DOI: 10.1093/humrep/det114] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
73
|
Pazaitou-Panayiotou K, Chemonidou C, Poupi A, Koureta M, Kaprara A, Lambropoulou M, Constantinidis TC, Galaktidou G, Koffa M, Kiziridou A, Kakolyris S, Kolios G, Kortsaris A, Chatzaki E. Gonadotropin-releasing hormone neuropeptides and receptor in human breast cancer: correlation to poor prognosis parameters. Peptides 2013; 42:15-24. [PMID: 23287110 DOI: 10.1016/j.peptides.2012.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/14/2012] [Accepted: 12/17/2012] [Indexed: 12/26/2022]
Abstract
Expression of the two gonadotropin-releasing hormone homologue peptides GnRHI and GnRHII and their receptor GnRHR has been demonstrated in a number of malignancies. In hormone-dependent breast cancer, GnRH analogs are used for therapy in premenopausal women. Gene expression of GnRHI, II and R was studied in breast biopsies from primary breast adenocarcinoma obtained from the tumor and the adjacent benign tissue. Levels were evaluated by a multiplex real-time RT-PCR. GnRHI transcripts were detected in 14.7% of the benign and 29.4% malignant biopsies and GnRHII in 21.2% benign and 44.1% malignant biopsies. GnRHR was also more frequent in the malignant (54.2%) than in the benign (24.0%) biopsies, at similar expression levels. No transcripts were detected in biopsies from healthy individuals. There was a strong correlation between the presence of GnRHI and GnRHII transcripts and their receptor in the benign and the malignant biopsies. GnRHI, II and R expression correlated significantly with poor prognosis pathological parameters. Immunohistochemistry for GnRHR revealed expression in malignant cells and in epithelial cells of mammary ducts of the adjacent area with pre-cancerous features. In contrast, GnRH I and II peptides were rarely expressed at low levels in breast cancer cells. In conclusion GnRH peptides and receptor are expressed more frequently in breast tumors than in the adjacent mammary tissue, representing a malignant feature. Their expression correlated to tumor characteristics of poor prognosis and was therefore related to more aggressive malignancies. Concomitant expression of peptides and receptor supports an autocrine/paracrine regulating role.
Collapse
|
74
|
Gadadhar S, Karande AA. Abrin immunotoxin: targeted cytotoxicity and intracellular trafficking pathway. PLoS One 2013; 8:e58304. [PMID: 23472175 PMCID: PMC3589266 DOI: 10.1371/journal.pone.0058304] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/01/2013] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Immunotherapy is fast emerging as one of the leading modes of treatment of cancer, in combination with chemotherapy and radiation. Use of immunotoxins, proteins bearing a cell-surface receptor-specific antibody conjugated to a toxin, enhances the efficacy of cancer treatment. The toxin Abrin, isolated from the Abrus precatorius plant, is a type II ribosome inactivating protein, has a catalytic efficiency higher than any other toxin belonging to this class of proteins but has not been exploited much for use in targeted therapy. METHODS Protein synthesis assay using (3)[H] L-leucine incorporation; construction and purification of immunotoxin; study of cell death using flow cytometry; confocal scanning microscopy and sub-cellular fractionation with immunoblot analysis of localization of proteins. RESULTS We used the recombinant A chain of abrin to conjugate to antibodies raised against the human gonadotropin releasing hormone receptor. The conjugate inhibited protein synthesis and also induced cell death specifically in cells expressing the receptor. The conjugate exhibited differences in the kinetics of inhibition of protein synthesis, in comparison to abrin, and this was attributed to differences in internalization and trafficking of the conjugate within the cells. Moreover, observations of sequestration of the A chain into the nucleus of cells treated with abrin but not in cells treated with the conjugate reveal a novel pathway for the movement of the conjugate in the cells. CONCLUSIONS This is one of the first reports on nuclear localization of abrin, a type II RIP. The immunotoxin mAb F1G4-rABRa-A, generated in our laboratory, inhibits protein synthesis specifically on cells expressing the gonadotropin releasing hormone receptor and the pathway of internalization of the protein is distinct from that seen for abrin.
Collapse
Affiliation(s)
- Sudarshan Gadadhar
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
| | - Anjali A. Karande
- Department of Biochemistry, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
75
|
Pharmacoperone IN3 enhances the apoptotic effect of leuprolide in prostate cancer cells by increasing the gonadotropin-releasing hormone receptor in the cell membrane. Anticancer Drugs 2013; 23:959-69. [PMID: 22713594 DOI: 10.1097/cad.0b013e328355ef88] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) agonists are widely used for the treatment of advanced prostate cancer (PCa). Agonists activate the GnRH receptor (GnRH-R), triggering apoptosis in PCa cells. In gonadotropes, the amount of GnRH-R in the plasma membrane is regulated by protein folding and endoplasmic reticulum retention, mechanisms that can be overcome by the pharmacoperone IN3. Our aim was to describe the intracellular distribution of GnRH-R in PCa cells and its relation to response to GnRH analog treatments. The expressions of GnRH-R in PCa biopsies were evaluated by immunohistochemistry and the intracellular distribution was determined by immunofluorescence in primary cell cultures from human PCa samples. Cultured cells were pretreated with IN3 and then with leuprolide. Cell survival was evaluated by 1-(4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT) thiazolyl blue formazan and cell cycle and apoptosis by flow cytometry. We observed that the expression of GnRH-R decreased according to malignant progression. Most GnRH-R are located inside the cell, colocalizing with endoplasmic reticulum markers. The treatment with IN3 decreased cellular GnRH-R retention, increasing plasma membrane expression in approximately 60%. Pretreatment with IN3 decreased PCa cell survival compared with leuprolide-alone treatment, primarily because of an increase in apoptosis. We conclude that the response of PCa cells to leuprolide is related to the amount of GnRH-R in the plasma membrane. Therefore, pretreatment evaluation of the amount of these receptors may be a predictor of the outcome of leuprolide treatment in PCa patients. Assessment of systemic IN3 effect would be necessary to determine its utility as an adjuvant treatment in hormone-resistant tumors.
Collapse
|
76
|
Lajkó E, Szabó I, Andódy K, Pungor A, Mező G, Kőhidai L. Investigation on chemotactic drug targeting (chemotaxis and adhesion) inducer effect of GnRH-III derivatives in Tetrahymena and human leukemia cell line. J Pept Sci 2012. [PMID: 23208929 DOI: 10.1002/psc.2472] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GnRH-III has been shown to exert a cytotoxic effect on the GnRH-R positive tumor cells. The chemotactic drug targeting (CDT) represents a new way for drug delivery approach based on selective chemoattractant guided targeting. The major goal of the present work was to develop and investigate various GnRH-III derivatives as potential targeting moieties for CDT. The cell physiological effects (chemotaxis, adhesion, and signaling) induced by three native GnRHs (hGnRH-I, cGnRH-II, and lGnRH-III) and nine GnRH-III derivatives were evaluated in two model cells (Tetrahymena pyriformis and Mono Mac 6 human monocytes). According to our results, the native GnRH-III elicited the highest chemoattractant and adhesion inducer activities of all synthesized peptides in micromolar concentrations in monocytes. With respect to chemoattraction, dimeric derivatives linked by a disulfide bridge ([GnRH-III(C)](2) ) proved to be efficient in both model cells; furthermore, acetylation of the linker region ([GnRH-III(Ac-C)](2) ) could slightly improve the chemotactic and adhesion effects in monocytes. The length of the peptide and the type of N-terminal amino acid could also determine the chemotactic and adhesion modulation potency of each fragment. The application of the chemoattractant GnRH-III derivatives was accompanied by a significant activation of phosphatidylinositol 3-kinase in both model cells. In summary, our work on low-level differentiated model cells of tumors has proved that GnRH-III and some of its synthetic derivatives are promising candidates to be applied in CDT: these compounds might act both as carrier, delivery unit, and antitumor agents.
Collapse
Affiliation(s)
- Eszter Lajkó
- Department of Genetics Cell and Immunobiology, Semmelweis University, Nagyvárad tér. 4, H-1089, Budapest, Hungary
| | | | | | | | | | | |
Collapse
|
77
|
Choi D. The consequences of mutations in the reproductive endocrine system. Dev Reprod 2012; 16:235-51. [PMID: 25949097 PMCID: PMC4282240 DOI: 10.12717/dr.2012.16.4.235] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 12/01/2012] [Accepted: 12/10/2012] [Indexed: 01/03/2023]
Abstract
The reproductive activity in male mammals is well known to be regulated by the hypothalamus-pituitary- gonad axis. The hypothalamic neurons secreting gonadotropin releasing hormone (GnRH) govern the reproductive neuroendocrine system by integrating all the exogenous information impinging on themselves. The GnRH synthesized and released from the hypothalamus arrives at the anterior pituitary through the portal vessels, provoking the production of the gonadotropins(follicle-stimulating hormone (FSH) and luteinizing hormone (LH)) at the same time. The gonadotropins affect the gonads to promote spermatogenesis and to secret testosterone. Testosterone acts on the GnRH neurons by a feedback loop through the circulatory system, resulting in the balance of all the hormones by regulating reproductive activities. These hormones exert their effects by acting on their own receptors, which are included in the signal transduction pathways as well. Unexpected aberrants are arised during this course of action of each hormone. This review summarizes these abnormal phenomena, including various mutations of molecules and their actions related to the reproductive function.
Collapse
Affiliation(s)
- Donchan Choi
- Dept. of Life Science, College of Environmental Sciences, Yong-In University, Yongin 449-714, Korea
| |
Collapse
|
78
|
Melamed P, Savulescu D, Lim S, Wijeweera A, Luo Z, Luo M, Pnueli L. Gonadotrophin-releasing hormone signalling downstream of calmodulin. J Neuroendocrinol 2012; 24:1463-75. [PMID: 22775470 DOI: 10.1111/j.1365-2826.2012.02359.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 06/24/2012] [Accepted: 07/03/2012] [Indexed: 01/26/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH) regulates reproduction via binding a G-protein coupled receptor on the surface of the gonadotroph, through which it transmits signals, mostly via the mitogen-activated protein (MAPK) cascade, to increase synthesis of the gonadotrophin hormones: luteinising hormone (LH) and follicle-stimulating hormone (FSH). Activation of the MAPK cascade requires an elevation in cytosolic Ca(2+) levels, which is a result of both calcium influx and mobilisation from intracellular stores. However, Ca(2+) also transmits signals via an MAPK-independent pathway, through binding calmodulin (CaM), which is then able to bind a number of proteins to impart diverse downstream effects. Although the ability of GnRH to activate CaM was recognised over 20 years ago, only recently have some of the downstream effects been elucidated. GnRH was shown to activate the CaM-dependent phosphatase, calcineurin, which targets gonadotrophin gene expression both directly and indirectly via transcription factors such as nuclear factor of activated T-cells and Nur77, the Transducer of Regulated CREB (TORC) co-activators and also the prolyl isomerase, Pin1. Gonadotrophin gene expression is also regulated by GnRH-induced CaM-dependent kinases (CaMKs); CaMKI is able to derepress the histone deacetylase-inhibition of β-subunit gene expression, whereas CaMKII appears to be essential for the GnRH-activation of all three subunit genes. Asides from activating gonadotrophin gene expression, GnRH also exerts additional effects on gonadotroph function, some of which clearly occur via CaM, including the proliferation of immature gonadotrophs, which is dependent on calcineurin. In this review, we summarise these pathways, and discuss the additional functions that have been proposed for CaM with respect to modifying GnRH-induced signalling pathways via the regulation of the small GTP-binding protein, Gem, and/or the regulator of G-protein signalling protein 2.
Collapse
Affiliation(s)
- P Melamed
- Technion-Israel Institute of Technology, Haifa, Israel.
| | | | | | | | | | | | | |
Collapse
|
79
|
Limonta P, Montagnani Marelli M, Mai S, Motta M, Martini L, Moretti RM. GnRH receptors in cancer: from cell biology to novel targeted therapeutic strategies. Endocr Rev 2012; 33:784-811. [PMID: 22778172 DOI: 10.1210/er.2012-1014] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The crucial role of pituitary GnRH receptors (GnRH-R) in the control of reproductive functions is well established. These receptors are the target of GnRH agonists (through receptor desensitization) and antagonists (through receptor blockade) for the treatment of steroid-dependent pathologies, including hormone-dependent tumors. It has also become increasingly clear that GnRH-R are expressed in cancer tissues, either related (i.e. prostate, breast, endometrial, and ovarian cancers) or unrelated (i.e. melanoma, glioblastoma, lung, and pancreatic cancers) to the reproductive system. In hormone-related tumors, GnRH-R appear to be expressed even when the tumor has escaped steroid dependence (such as castration-resistant prostate cancer). These receptors are coupled to a G(αi)-mediated intracellular signaling pathway. Activation of tumor GnRH-R by means of GnRH agonists elicits a strong antiproliferative, antimetastatic, and antiangiogenic (more recently demonstrated) activity. Interestingly, GnRH antagonists have also been shown to elicit a direct antitumor effect; thus, these compounds behave as antagonists of GnRH-R at the pituitary level and as agonists of the same receptors expressed in tumors. According to the ligand-induced selective-signaling theory, GnRH-R might assume various conformations, endowed with different activities for GnRH analogs and with different intracellular signaling pathways, according to the cell context. Based on these consistent experimental observations, tumor GnRH-R are now considered a very interesting candidate for novel molecular, GnRH analog-based, targeted strategies for the treatment of tumors expressing these receptors. These agents include GnRH agonists and antagonists, GnRH analog-based cytotoxic (i.e. doxorubicin) or nutraceutic (i.e. curcumin) hybrids, and GnRH-R-targeted nanoparticles delivering anticancer compounds.
Collapse
Affiliation(s)
- Patrizia Limonta
- Section of Biomedicine and Endocrinology, Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy.
| | | | | | | | | | | |
Collapse
|
80
|
Tan O, Carr BR, Beshay VE, Bukulmez O. The extrapituitary effects of GnRH antagonists and their potential clinical implications: a narrated review. Reprod Sci 2012; 20:16-25. [PMID: 23012318 DOI: 10.1177/1933719112459244] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Potential roles of gonadotropin-releasing hormone (GnRH) antagonists on GnRH/GnRH receptor systems and their effects on the extrapituitary tissues are largely elusive. In this narrated review, we summarized the systemic effects of GnRH antagonists on ovary, endometrium, embryo implantation, placental development, fetal teratogenicity, reproductive tissue cancer cells, and heart while briefly reviewing the GnRH and GnRH receptor system. GnRH antagonists may have direct effects on ovarian granulosa cells. Data are conflicting regarding their effects on endometrial receptivity. The GnRH antagonists may potentially have detrimental effect on early placentation by decreasing the invasive ability of cytotrophoblasts if the exposure to them occurs during early pregnancy. The GnRH antagonists were not found to increase the rates of congenital malformations. Comparative clinical data are required to explore their systemic effects on various extrapituitary tissues such as on cardiac function in the long term as well as their potential use in other human cancers that express GnRH receptors.
Collapse
Affiliation(s)
- Orkun Tan
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | | | | | |
Collapse
|
81
|
Gianetti E, Hall JE, Au MG, Kaiser UB, Quinton R, Stewart JA, Metzger DL, Pitteloud N, Mericq V, Merino PM, Levitsky LL, Izatt L, Lang-Muritano M, Fujimoto VY, Dluhy RG, Chase ML, Crowley WF, Plummer L, Seminara SB. When genetic load does not correlate with phenotypic spectrum: lessons from the GnRH receptor (GNRHR). J Clin Endocrinol Metab 2012; 97:E1798-807. [PMID: 22745237 PMCID: PMC3431570 DOI: 10.1210/jc.2012-1264] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CONTEXT A broad spectrum of GnRH-deficient phenotypes has been identified in individuals with both mono- and biallelic GNRHR mutations. OBJECTIVE The objective of the study was to determine the correlation between the severity of the reproductive phenotype(s) and the number and functional severity of rare sequence variants in GNRHR. SUBJECTS Eight hundred sixty-three probands with different forms of GnRH deficiency, 46 family members and 422 controls were screened for GNRHR mutations. The 70 subjects (32 patients and 38 family members) harboring mutations were divided into four groups (G1-G4) based on the functional severity of the mutations (complete or partial loss of function) and the number of affected alleles (monoallelic or biallelic) with mutations, and these classes were mapped on their clinical phenotypes. RESULTS The prevalence of heterozygous rare sequence variants in GNRHR was significantly higher in probands vs. controls (P < 0.01). Among the G1-G3 groups (homozygous subjects with successively decreasing severity and number of mutations), the hypogonadotropic phenotype related to their genetic load. In contrast, subjects in G4, with only monoallelic mutations, demonstrated a greater diversity of clinical phenotypes. CONCLUSIONS In patients with GnRH deficiency and biallelic mutations in GNRHR, genetic burden defined by severity and dose is associated with clinical phenotype. In contrast, for patients with monoallelic GNRHR mutations this correlation does not hold. Taken together, these data indicate that as-yet-unidentified genetic and/or environmental factors may combine with singly mutated GNRHR alleles to produce reproductive phenotypes.
Collapse
Affiliation(s)
- Elena Gianetti
- Harvard Center for Reproductive Sciences and Reproductive Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, BHX 504, Boston, Massachusetts 02114, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Lainas GT, Kolibianakis EM, Sfontouris IA, Zorzovilis IZ, Petsas GK, Tarlatzi TB, Tarlatzis BC, Lainas TG. Outpatient management of severe early OHSS by administration of GnRH antagonist in the luteal phase: an observational cohort study. Reprod Biol Endocrinol 2012; 10:69. [PMID: 22938051 PMCID: PMC3489837 DOI: 10.1186/1477-7827-10-69] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 08/27/2012] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Management of established severe OHSS requires prolonged hospitalization, occasionally in intensive care units, accompanied by multiple ascites punctures, correction of intravascular fluid volume and electrolyte imbalance. The aim of the present study was to evaluate whether it is feasible to manage women with severe OHSS as outpatients by treating them with GnRH antagonists in the luteal phase. METHODS This is a single-centre, prospective, observational, cohort study. Forty patients diagnosed with severe OHSS, five days post oocyte retrieval, were managed as outpatients after administration of GnRH antagonist (0.25 mg) daily from days 5 to 8 post oocyte retrieval, combined with cryopreservation of all embryos. The primary outcome measure was the proportion of patients with severe OHSS, in whom outpatient management was not feasible. RESULTS 11.3% (95% CI 8.3%-15.0%) of patients (40/353) developed severe early OHSS. None of the 40 patients required hospitalization following luteal antagonist administration and embryo cryopreservation. Ovarian volume, ascites, hematocrit, WBC, serum oestradiol and progesterone decreased significantly (P < 0.001) by the end of the monitoring period, indicating rapid resolution of severe OHSS. CONCLUSIONS The current study suggests, for the first time, that successful outpatient management of severe OHSS with antagonist treatment in the luteal phase is feasible and is associated with rapid regression of the syndrome, challenging the dogma of inpatient management. The proposed management is a flexible approach that minimizes unnecessary embryo transfer cancellations in the majority (88.7%) of high risk for OHSS patients.
Collapse
Affiliation(s)
- George T Lainas
- Eugonia Assisted Reproduction Unit, 7 Ventiri Street, 11528, Athens, Greece
| | - Efstratios M Kolibianakis
- Unit for Human Reproduction, 1st Department of Obstetrics & Gynecology, Papageorgiou General Hospital, Medical School, Aristotle University of Thessaloniki, Ring Road, Nea Efkarpia, 56429, Thessaloniki, Greece
| | | | | | - George K Petsas
- Eugonia Assisted Reproduction Unit, 7 Ventiri Street, 11528, Athens, Greece
| | - Theoni B Tarlatzi
- Unit for Human Reproduction, 1st Department of Obstetrics & Gynecology, Papageorgiou General Hospital, Medical School, Aristotle University of Thessaloniki, Ring Road, Nea Efkarpia, 56429, Thessaloniki, Greece
| | - Basil C Tarlatzis
- Unit for Human Reproduction, 1st Department of Obstetrics & Gynecology, Papageorgiou General Hospital, Medical School, Aristotle University of Thessaloniki, Ring Road, Nea Efkarpia, 56429, Thessaloniki, Greece
| | - Trifon G Lainas
- Eugonia Assisted Reproduction Unit, 7 Ventiri Street, 11528, Athens, Greece
| |
Collapse
|
83
|
Choi SG, Jia J, Pfeffer RL, Sealfon SC. G proteins and autocrine signaling differentially regulate gonadotropin subunit expression in pituitary gonadotrope. J Biol Chem 2012; 287:21550-60. [PMID: 22549790 DOI: 10.1074/jbc.m112.348607] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) acts at gonadotropes to direct the synthesis of the gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH). The frequency of GnRH pulses determines the pattern of gonadotropin synthesis. Several hypotheses for how the gonadotrope decodes GnRH frequency to regulate gonadotropin subunit genes differentially have been proposed. However, key regulators and underlying mechanisms remain uncertain. We investigated the role of individual G proteins by perturbations using siRNA or bacterial toxins. In LβT2 gonadotrope cells, FSHβ gene induction depended predominantly on Gα(q/11), whereas LHβ expression depended on Gα(s). Specifically reducing Gα(s) signaling also disinhibited FSHβ expression, suggesting the presence of a Gα(s)-dependent signal that suppressed FSH biosynthesis. The presence of secreted factors influencing FSHβ expression levels was tested by studying the effects of conditioned media from Gα(s) knockdown and cholera toxin-treated cells on FSHβ expression. These studies and related Transwell culture experiments implicate Gα(s)-dependent secreted factors in regulating both FSHβ and LHβ gene expression. siRNA studies identify inhibinα as a Gα(s)-dependent GnRH-induced autocrine regulatory factor that contributes to feedback suppression of FSHβ expression. These results uncover differential regulation of the gonadotropin genes by Gα(q/11) and by Gα(s) and implicate autocrine and gonadotrope-gonadotrope paracrine regulatory loops in the differential induction of gonadotropin genes.
Collapse
Affiliation(s)
- Soon-Gang Choi
- Center for Translational Systems Biology and the Department of Neurology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | |
Collapse
|
84
|
Cota AMM, Oliveira JBA, Petersen CG, Mauri AL, Massaro FC, Silva LFI, Nicoletti A, Cavagna M, Baruffi RLR, Franco JG. GnRH agonist versus GnRH antagonist in assisted reproduction cycles: oocyte morphology. Reprod Biol Endocrinol 2012; 10:33. [PMID: 22540993 PMCID: PMC3464873 DOI: 10.1186/1477-7827-10-33] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/27/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The selection of developmentally competent human gametes may increase the efficiency of assisted reproduction. Spermatozoa and oocytes are usually assessed according to morphological criteria. Oocyte morphology can be affected by the age, genetic characteristics, and factors related to controlled ovarian stimulation. However, there is a lack of evidence in the literature concerning the effect of gonadotropin-releasing hormone (GnRH) analogues, either agonists or antagonists, on oocyte morphology. The aim of this randomized study was to investigate whether the prevalence of oocyte dysmorphism is influenced by the type of pituitary suppression used in ovarian stimulation. METHODS A total of 64 patients in the first intracytoplasmic sperm injection (ICSI) cycle were prospectively randomized to receive treatment with either a GnRH agonist with a long-term protocol (n: 32) or a GnRH antagonist with a multi-dose protocol (n: 32). Before being subjected to ICSI, the oocytes at metaphase II from both groups were morphologically analyzed under an inverted light microscope at 400x magnification. The oocytes were classified as follows: normal or with cytoplasmic dysmorphism, extracytoplasmic dysmorphism, or both. The number of dysmorphic oocytes per total number of oocytes was analyzed. RESULTS Out of a total of 681 oocytes, 189 (27.8%) were morphologically normal, 220 (32.3%) showed cytoplasmic dysmorphism, 124 (18.2%) showed extracytoplasmic alterations, and 148 (21.7%) exhibited both types of dysmorphism. No significant difference in oocyte dysmorphism was observed between the agonist- and antagonist-treated groups (P>0.05). Analysis for each dysmorphism revealed that the most common conditions were alterations in polar body shape (31.3%) and the presence of diffuse cytoplasmic granulations (22.8%), refractile bodies (18.5%) and central cytoplasmic granulations (13.6%). There was no significant difference among individual oocyte dysmorphisms in the agonist- and antagonist-treated groups (P>0.05). CONCLUSIONS Our randomized data indicate that in terms of the quality of oocyte morphology, there is no difference between the antagonist multi-dose protocol and the long-term agonist protocol. If a GnRH analogue used for pituitary suppression in IVF cycles influences the prevalence of oocyte dysmorphisms, there does not appear to be a difference between the use of an agonist as opposed to an antagonist.
Collapse
Affiliation(s)
- Ana Marcia M Cota
- Department of Gynecology and Obstetrics, Botucatu Medical School, São Paulo State University—UNESP, Botucatu, Brazil
| | - Joao Batista A Oliveira
- Department of Gynecology and Obstetrics, Botucatu Medical School, São Paulo State University—UNESP, Botucatu, Brazil
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - Claudia G Petersen
- Department of Gynecology and Obstetrics, Botucatu Medical School, São Paulo State University—UNESP, Botucatu, Brazil
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - Ana L Mauri
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - Fabiana C Massaro
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - Liliane FI Silva
- Department of Gynecology and Obstetrics, Botucatu Medical School, São Paulo State University—UNESP, Botucatu, Brazil
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - Andreia Nicoletti
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
| | - Mario Cavagna
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
- Women’s Health Reference Center, Perola Byington Hospital, Sao Paulo, Brazil
| | - Ricardo LR Baruffi
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| | - José G Franco
- Department of Gynecology and Obstetrics, Botucatu Medical School, São Paulo State University—UNESP, Botucatu, Brazil
- Center for Human Reproduction Prof. Franco Junior, Ribeirao Preto, Brazil
- Paulista Center for Diagnosis, Research and Training, Ribeirao Preto, Brazil
| |
Collapse
|
85
|
Ling Poon S, Lau MT, Hammond GL, Leung PCK. Gonadotropin-releasing hormone-II increases membrane type I metalloproteinase production via beta-catenin signaling in ovarian cancer cells. Endocrinology 2011; 152:764-72. [PMID: 21239435 DOI: 10.1210/en.2010-0942] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH-II is produced by ovarian cancer cells and enhances their invasiveness in vitro. In our studies of OVCAR-3 and CaOV-3 ovarian cancer cell lines, GnRH-II treatment induced phosphorylation of Akt and glycogen synthase kinase (GSK)3β, as well as β-catenin accumulation in the nucleus, and the latter was reduced by small interfering RNA (siRNA)-mediated depletion of the GnRH receptor. The phosphatidylinositol 3 kinase (PI3K)/Akt pathway is involved in β-catenin-dependent signaling, and pretreatment of these human ovarian cancer cells with a PI3K/Akt inhibitor, LY294002, attenuated GnRH-II-stimulated phosphorylation of GSK3β and inhibited GnRH-II-induced invasion. It also attenuated GnRH-II induced trans-activation of a β-catenin-dependent reporter gene, most likely because GSK3β phosphorylation promotes translocation of β-catenin to the nucleus. Membrane type I matrix metalloproteinase (MT1-MMP) contributes to tumor progression directly, or by processing the latent MMP-2 zymogen, and is a known target of β-catenin signaling. When OVCAR-3 and CaOV-3 cells were treated with GnRH-II, MT1-MMP levels increased approximately 3-fold, whereas siRNA-mediated depletion of GnRH receptor or pretreatment with LY294002 abrogated this. In addition, lithium chloride, which increases GSK3β phosphorylation and the nuclear translocation of β-catenin, increased MT1-MMP levels in these ovarian cancer cells. By contrast, depletion of β-catenin by siRNA treatment abolished GnRH-II-induced MT1-MMP synthesis and reduced their invasive potential. Furthermore, siRNA-mediated reduction of MT1-MMP levels reduced GnRH-II-induced invasion in ovarian cancer cells. We therefore conclude that GnRH-II stimulates the PI3K/Akt pathway, and the phosphorylation of GSK3β, thereby enhancing the β-catenin-dependent up-regulation of MT1-MMP production, which contributes to ovarian cancer metastasis.
Collapse
Affiliation(s)
- Song Ling Poon
- Department of Obstetrics and Gynaecology, University of British Columbia, Room 2H-30, 4490 Oak Street, Vancouver, British Columbia, Canada V6H 3V5
| | | | | | | |
Collapse
|
86
|
Cheung LWT, Mak ASC, Cheung ANY, Ngan HYS, Leung PCK, Wong AST. P-cadherin cooperates with insulin-like growth factor-1 receptor to promote metastatic signaling of gonadotropin-releasing hormone in ovarian cancer via p120 catenin. Oncogene 2011; 30:2964-74. [PMID: 21317933 DOI: 10.1038/onc.2011.7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) is a potent prometastatic factor in ovarian cancer, but the intracellular signaling events are not well understood. The classical Gα(q)-phospholipase C signal transduction pathway known to operate in the pituitary is not involved in GnRH actions at non-pituitary targets. Here we showed that GnRH treatment of ovarian cancer cells led to a rapid and remarkable tyrosine phosphorylation of p120 catenin (p120(ctn)), which was mediated by P-cadherin. The use of P-cadherin small interfering RNA or neutralizing antibodies to inhibit P-cadherin expression and function resulted in diminished p120(ctn) activation, confirming that the effect was P-cadherin specific. On exploring how P-cadherin, which lacks intrinsic kinase activity, might regulate the activation of p120(ctn), we found that P-cadherin could induce the ligand-independent activation of insulin-like growth factor-1 receptor (IGF-1R). Inhibition of IGF-1R expression or its activity significantly inhibited GnRH-induced p120(ctn) activation, and the subsequent cell migration and invasion. In addition, we showed that IGF-1R regulation by P-cadherin was associated with complex formation between IGF-1R and P-cadherin, and this regulation was also observed to be in vivo correlated with metastasis. Furthermore, using a mouse model of ovarian cancer metastasis, GnRH receptor knockdown was shown to diminish peritoneal dissemination of tumors and ascites formation. These findings suggest for the first time that GnRH can initiate an outside-in p120(ctn) signal transduction through the cross-talk between P-cadherin and IGF-1R, thus providing a novel molecular mechanism by which GnRH may control the high level of aggressiveness and invasion and metastasis potential that are characteristic of ovarian cancer.
Collapse
Affiliation(s)
- L W T Cheung
- School of Biological Sciences, University of Hong Kong, Hong Kong, Hong Kong
| | | | | | | | | | | |
Collapse
|
87
|
Quintanar JL, Salinas E, Quintanar-Stephano A. Gonadotropin-releasing hormone reduces the severity of experimental autoimmune encephalomyelitis, a model of multiple sclerosis. Neuropeptides 2011; 45:43-8. [PMID: 21056467 DOI: 10.1016/j.npep.2010.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 09/09/2010] [Accepted: 10/14/2010] [Indexed: 10/18/2022]
Abstract
It has been reported that the spinal cord possesses Gonadotropin-releasing hormone (GnRH) receptor and that GnRH has neurotrophic properties. Experimental autoimmune encephalomyelitis (EAE) causes neurodegeneration in spinal cord. Thus, the present study was designed to determine whether administration of GnRH reduces the severity of EAE. The clinical signs of locomotion, axonal morphometry and neurofilaments (NFs) expression were evaluated. Clinical signs remained significantly lower in EAE rats with GnRH administration compared to animals without treatment. Morphometric analysis, there were more axons of larger areas in the spinal cord of EAE+GnRH group compared to EAE animals. Western blot analysis demonstrated that GnRH administration significantly increased the expression of NFs of 68, 160 and 200kDa in the spinal cord of EAE animals. Our results indicate that GnRH administration reduces the severity of EAE in the rat.
Collapse
Affiliation(s)
- J Luis Quintanar
- Laboratory of Neurophysiology, Department of Physiology and Pharmacology, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad 940, C.P. 20131 Aguascalientes, México.
| | | | | |
Collapse
|
88
|
Zerani M, Catone G, Quassinti L, Maccari E, Bramucci M, Gobbetti A, Maranesi M, Boiti C, Parillo F. In vitro effects of gonadotropin-releasing hormone (GnRH) on Leydig cells of adult alpaca (Lama pacos) testis: GnRH receptor immunolocalization, testosterone and prostaglandin synthesis, and cyclooxygenase activities. Domest Anim Endocrinol 2011; 40:51-9. [PMID: 20961724 DOI: 10.1016/j.domaniend.2010.08.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 08/28/2010] [Accepted: 08/29/2010] [Indexed: 10/19/2022]
Abstract
The main objective of this study was to examine the modulatory in vitro effects of gonadotropin-releasing hormone (GnRH) on isolated Leydig cells of adult alpaca (Lama pacos) testis. We first evaluated the presence of GnRH receptor (GnRHR) and cyclooxygenase (COX) 1 and COX2 in alpaca testis. We then studied the in vitro effects of buserelin (GnRH analogue), antide (GnRH antagonist), and buserelin plus antide or inhibitor of phospholipase C (compound 48/80) and COXs (acetylsalicylic acid) on the production of testosterone, PGE(2), and PGF(2α) and on the enzymatic activities of COX1 and COX2. Immunoreactivity for GnRHR was detected in the cytoplasm of Leydig cells and in the acrosomal region of spermatids. COX1 and COX2 immunosignals were noted in the cytoplasm of spermatogonia, spermatocytes, spermatids, Leydig cells, and Sertoli cells. Western blot analysis confirmed the GnRHR and COX1 presence in alpaca testis. The in vitro experiments showed that buserelin alone increased (P < 0.01) and antide and buserelin plus acetylsalicylic acid decreased (P < 0.01) testosterone and PGF(2α) production and COX1 activity, whereas antide and compound 48/80 counteracted buserelin effects. Prostaglandin E(2) production and COX2 activity were not affected by buserelin or antide. These data suggest that GnRH directly up-regulates testosterone production in Leydig cells of adult alpaca testis with a postreceptorial mechanism that involves PLC, COX1, and PGF(2α).
Collapse
Affiliation(s)
- M Zerani
- Scuola di Scienze Mediche Veterinarie, Università di Camerino, I-62024 Matelica, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Poon SL, Klausen C, Hammond GL, Leung PCK. 37-kDa laminin receptor precursor mediates GnRH-II-induced MMP-2 expression and invasiveness in ovarian cancer cells. Mol Endocrinol 2010; 25:327-38. [PMID: 21193558 DOI: 10.1210/me.2010-0334] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH-II enhances ovarian cancer cell invasion in an autocrine manner. We have now found that GnRH-II increases 37-kDa laminin receptor precursor (LRP) production in GnRH receptor (GnRHR)-positive OVCAR-3 and CaOV-3 ovarian cancer cells, while small interfering RNA (siRNA)-mediated depletion of GnRH-II or GnRHR mRNA abrogates this. The invasiveness of ovarian cancer cells is also reduced >85% by siRNA-mediated knockdown of LRP levels and >50% by pretreatment of Matrigel with a synthetic peptide that blocks interactions between laminin and the 67-kDa nonintegrin laminin receptor which comprises two LRP subunits. Conversely, overexpressing LRP in CaOV-3 cells increases their invasiveness 5-fold, while overexpressing LRP with a nonfunctional laminin-binding site does not. Depletion of LRP by siRNA treatment reduces CaOV-3 cell attachment to laminin-coated plates by ∼80% but only reduces their binding to Matrigel by ∼20%. Thus, while LRP influences CaOV-3 cell adhesion to laminin, LRP must act in other ways to enhance invasion. Matrix metalloproteinases (MMPs) are key mediators of invasion, and LRP siRNA treatment of OVCAR-3 and CaOV-3 cells inhibits MMP-2 but not MMP-9 mRNA levels. Overexpressing LRP in these cells increases MMP-2 production specifically, while a laminin-binding deficient LRP does not. Importantly, LRP siRNA treatment abolishes GnRH-II-induced MMP-2 production, and invasion in OVCAR-3 and CaOV-3 cells, which was also seen after MMP-2 siRNA treatment. These results suggest that GnRH-II-induced LRP expression increases the amount of the 67-kDa nonintegrin laminin receptor, which appears to interact with laminin in the extracellular matrix to promote MMP-2 expression and enhance ovarian cancer cell invasion.
Collapse
Affiliation(s)
- Song Ling Poon
- Department of Obstetrics and Gynecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
90
|
Russo A, Castiglione F, Salonia A, Benigni F, Rigatti P, Montorsi F, Andersson KE, Hedlund P. Effects of the gonadotropin-releasing hormone antagonist ganirelix on normal micturition and prostaglandin E(2)-induced detrusor overactivity in conscious female rats. Eur Urol 2010; 59:868-74. [PMID: 21196074 DOI: 10.1016/j.eururo.2010.12.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2010] [Accepted: 12/10/2010] [Indexed: 01/15/2023]
Abstract
BACKGROUND Gonadotropin-releasing hormone (GnRH) antagonists have been reported to have beneficial effects on lower urinary tract symptoms in patients with benign prostatic hyperplasia. OBJECTIVE Our aim was to investigate the effects of ganirelix, a GnRH receptor antagonist, on bladder function and detrusor overactivity (DO) in female rats. DESIGN, SETTING, AND PARTICIPANTS Female Sprague-Dawley rats received 2 wk of daily systemic (0.1 mg/kg) or acute intravesical administration (IVES; 0.14 mg/l or 1.4 mg/l) ganirelix or vehicle (controls). MEASUREMENTS Assessments were obtained using cystometry in awake rats, organ bath studies, enzyme-linked immunosorbent assay, and western blot (WB). RESULTS AND LIMITATIONS Luteinising hormone levels were lower in rats treated systemically with ganirelix than in controls. No differences were observed in body or bladder weights. Micturition interval (MI), micturition volume (MV), residual volume, and bladder capacity (BC) were similar in both groups at baseline. No differences in urodynamic pressure parameters were observed between groups at baseline. Intravesical prostaglandin E(2) reduced MI, MV, and BC, and it increased basal pressure (BP), threshold pressure (TP), flow pressure (FP), and maximum pressure (MP) in all rats. MI, MV, and BC were reduced by 43%±4%, 50%±4%, and 43%±4% (controls) versus 22%±3%, 23%±3%, and 21%±3% (ganirelix-treated rats; p<0.001). TP and FP increased by 38%±8% and 30%±4% (controls) versus 16%±7% and 16%±5% (ganirelix; p<0.05). The maximal force of contractions for carbachol was larger in detrusor from ganirelix-treated rats (231% vs 177% of 60 mM K+-induced contractions). At 0.14 mg/l, but not 0.14 mg/l, IVES ganirelix increased MI, MV, and BC and decreased BP, TP, FP, and MP. In vitro, ganirelix had no effect on detrusor function. The gonadotropin-releasing hormone receptor was expressed (by WB) in the bladder mucosa. CONCLUSIONS Systemic treatment with ganirelix counteracted experimental DO in female rats. Because bladder preparations from these rats exhibited larger contractions to carbachol and because intravesical ganirelix affected both micturition intervals and urodynamic pressure profiles, a peripheral site of action of ganirelix in the urinary bladder cannot be excluded.
Collapse
Affiliation(s)
- Andrea Russo
- Urological Research Institute, San Raffaele University, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
91
|
Siejka A, Schally AV, Block NL, Barabutis N. Mechanisms of inhibition of human benign prostatic hyperplasia in vitro by the luteinizing hormone-releasing hormone antagonist cetrorelix. BJU Int 2010; 106:1382-8. [PMID: 20151966 DOI: 10.1111/j.1464-410x.2010.09215.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To assess the mechanism by which the luteinizing hormone-releasing hormone (LHRH) antagonist cetrorelix exerts its effects in men with benign prostatic hyperplasia (BPH), as it produces a long-lasting improvement in lower urinary tract symptoms that is only partly accounted for by the transient reduction in testosterone levels, and the beneficial results could be due to direct inhibitory effects of cetrorelix on the prostate exerted through prostatic LHRH receptors. MATERIALS AND METHODS Using the BPH-1 cell line we evaluated the effects of cetrorelix in vitro on the proliferation and the expression of receptors for LHRH, epidermal growth factor (EGF), α(1A) -adrenergic receptor, STAT-3 transcription factor and the response to growth factors insulin-like growth factor (IGF)-1 and -II and fibroblast growth factor (FGF)-2. RESULTS There was expression of LHRH receptors in the human BPH-1 cell line. Cetrorelix had inhibitory effects on the proliferation rate of BPH-1 cells, also reflected by the decrease in the expression of the proliferating cell nuclear antigen (PCNA). Cetrorelix inhibited the stimulatory effect of the growth factors IGF-I and -II and FGF-2 on the proliferation of this line. Cetrorelix also downregulated the expression of the receptors for LHRH and EGF, as well as of α(1A) -adrenergic receptors, and inhibited the activation of the STAT3 transcription factor. CONCLUSIONS The results show that in vitro cetrorelix can directly inhibit the proliferation rate of the human BPH-1 cell line by counteracting growth factors like IGF-I and -II and FGF-2, and downregulating the LHRH receptor and α-adrenergic receptors, as well as transcription factors.
Collapse
Affiliation(s)
- Agnieszka Siejka
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL, USA
| | | | | | | |
Collapse
|
92
|
Wang L, Chadwick W, Park SS, Zhou Y, Silver N, Martin B, Maudsley S. Gonadotropin-releasing hormone receptor system: modulatory role in aging and neurodegeneration. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2010; 9:651-60. [PMID: 20632963 PMCID: PMC2967575 DOI: 10.2174/187152710793361559] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2010] [Accepted: 02/25/2010] [Indexed: 12/15/2022]
Abstract
Receptors for hormones of the hypothalamic-pituitary-gonadal axis are expressed throughout the brain. Age-related decline in gonadal reproductive hormones cause imbalances of this axis and many hormones in this axis have been functionally linked to neurodegenerative pathophysiology. Gonadotropin-releasing hormone (GnRH) plays a vital role in both central and peripheral reproductive regulation. GnRH has historically been known as a pituitary hormone; however, in the past few years, interest has been raised in GnRH actions at non-pituitary peripheral targets. GnRH ligands and receptors are found throughout the brain where they may act to control multiple higher functions such as learning and memory function and feeding behavior. The actions of GnRH in mammals are mediated by the activation of a unique rhodopsin-like G protein-coupled receptor that does not possess a cytoplasmic carboxyl terminal sequence. Activation of this receptor appears to mediate a wide variety of signaling mechanisms that show diversity in different tissues. Epidemiological support for a role of GnRH in central functions is evidenced by a reduction in neurodegenerative disease after GnRH agonist therapy. It has previously been considered that these effects were not via direct GnRH action in the brain, however recent data has pointed to a direct central action of these ligands outside the pituitary. We have therefore summarized the evidence supporting a central direct role of GnRH ligands and receptors in controlling central nervous physiology and pathophysiology.
Collapse
Affiliation(s)
- Liyun Wang
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Wayne Chadwick
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Soo-Sung Park
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Yu Zhou
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Nathan Silver
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Bronwen Martin
- Metabolism Unit, Laboratory of Clinical Investigation, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging, National Institutes of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore MD 21224
| |
Collapse
|
93
|
Fontaine E, Fontbonne A. Clinical Use of GnRH Agonists in Canine and Feline Species. Reprod Domest Anim 2010; 46:344-53. [DOI: 10.1111/j.1439-0531.2010.01705.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
94
|
Mutated genes, pathways and processes in tumours. EMBO Rep 2010; 11:805-10. [PMID: 20847737 PMCID: PMC2948187 DOI: 10.1038/embor.2010.133] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 06/10/2010] [Accepted: 07/27/2010] [Indexed: 02/01/2023] Open
Abstract
This analysis of genes mutated in cancers, drawn from small and large-scale genome studies, allows the authors to draw a map of pathways and processes altered by cancers, revealing both common and specific changes. Integration of the many available sources of cancer gene information—such as large-scale tumour-resequencing studies— identifies the ‘usual suspect' genes, mutated in many tumour types, as well as different sets of mutated genes according to the specific tumour type. Scaling-up the analysis reveals that this large collection of mutated genes cluster into a smaller number of signalling pathways and processes. From this, we draw a map of the altered processes, and their combinations, in more than 10 tumours types. Literature searches identify pathways and processes that are covered sparsely in the literature, and invite the proposal of new hypotheses to investigate cancer initiation and progression.
Collapse
|
95
|
Bliss SP, Navratil AM, Xie J, Roberson MS. GnRH signaling, the gonadotrope and endocrine control of fertility. Front Neuroendocrinol 2010; 31:322-40. [PMID: 20451543 PMCID: PMC2923852 DOI: 10.1016/j.yfrne.2010.04.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2010] [Revised: 04/26/2010] [Accepted: 04/30/2010] [Indexed: 11/28/2022]
Abstract
Mammalian reproductive cycles are controlled by an intricate interplay between the hypothalamus, pituitary and gonads. Central to the function of this axis is the ability of the pituitary gonadotrope to appropriately respond to stimulation by gonadotropin-releasing hormone (GnRH). This review focuses on the role of cell signaling and in particular, mitogen-activated protein kinase (MAPK) activities regulated by GnRH that are necessary for normal fertility. Recently, new mouse models making use of conditional gene deletion have shed new light on the relationships between GnRH signaling and fertility in both male and female mice. Within the reproductive axis, GnRH signaling is initiated through discrete membrane compartments in which the receptor resides leading to the activation of the extracellular signal-regulated kinases (ERKs 1/2). As defined by gonadotrope-derived cellular models, the ERKs appear to play a central role in the regulation of a cohort of immediate early genes that regulate the expression of late genes that, in part, define the differentiated character of the gonadotrope. Recent data would suggest that in vivo, conditional, pituitary-specific disruption of ERK signaling by GnRH leads to a gender-specific perturbation of fertility. Double ERK knockout in the anterior pituitary leads to female infertility due to LH biosynthesis deficiency and a failure in ovulation. In contrast, male mice are modestly LH deficient; however, this does not have an appreciable impact on fertility.
Collapse
Affiliation(s)
- Stuart P Bliss
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States
| | | | | | | |
Collapse
|
96
|
Zhang X, Bocca S, Franchi A, Anderson S, Kaur M, Bajic VB, Oehninger S. Do GnRH analogues directly affect human endometrial epithelial cell gene expression? Mol Hum Reprod 2010; 16:347-60. [DOI: 10.1093/molehr/gaq012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
97
|
Cadherin switching and activation of p120 catenin signaling are mediators of gonadotropin-releasing hormone to promote tumor cell migration and invasion in ovarian cancer. Oncogene 2010; 29:2427-40. [PMID: 20118984 DOI: 10.1038/onc.2009.523] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) receptor expression is often elevated in ovarian cancer, but its potential role in ovarian cancer metastasis has just begun to be revealed. Cadherin switching is a crucial step during tumorigenesis, particularly in metastasis. Here, we showed that GnRH is an inducer of E- to P-cadherin switching, which is reminiscent of that seen during ovarian tumor progression. Overexpression of P-cadherin significantly enhanced, whereas knockdown of P-cadherin reduced migration and invasion regardless of E-cadherin expression, suggesting that inappropriate expression of P-cadherin contributes to the invasive phenotype. These effects of P-cadherin were mediated by activation of the Rho GTPases, Rac1, and Cdc42, through accumulation of p120 catenin (p120(ctn)) in the cytoplasm. The use of p120(ctn) small interfering RNA or chimeric cadherin construct to inhibit p120(ctn) expression and cytoplasmic localization, respectively, resulted in significant inhibition of cell migration and invasion, with a concomitant reduction in Rac1 and Cdc42 activation, confirming that the effect was p120(ctn) specific. Similarly, the migratory/invasive phenotype could be reversed by expression of dominant-negative Rac1 and Cdc42. These results identify for the first time cadherin switching and p120(ctn) signaling as important targets of GnRH function and as novel mediators of invasiveness and tumor progression in ovarian cancer.
Collapse
|
98
|
De Paul AL, Attademo AM, Carón RW, Soaje M, Torres AI, Jahn GA, Celis ME. Neuropeptide glutamic-isoleucine (NEI) specifically stimulates the secretory activity of gonadotrophs in primary cultures of female rat pituitary cells. Peptides 2009; 30:2081-7. [PMID: 19729046 DOI: 10.1016/j.peptides.2009.08.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 08/14/2009] [Accepted: 08/25/2009] [Indexed: 11/17/2022]
Abstract
The neuropeptide EI (NEI) is derived from proMCH. It activates GnRH neurons, and has been shown to stimulate the LH release following intracerebroventricular administration in several experimental models. The aim of the present paper was to evaluate NEI actions on pituitary hormone secretion and cell morphology in vitro. Pituitary cells from female rats were treated with NEI for a wide range of concentrations (1-400x10(-8)M) and time periods (1-5h). The media were collected and LH, FSH, PRL, and GH measured by RIA. The interaction between NEI (1, 10 and 100x10(-8)M) and GnRH (0.1 and 1x10(-9)M) was also tested. Pituitary cells were harvested for electron microscopy, and the immunogold immunocytochemistry of LH was assayed after 2 and 4h of NEI incubation. NEI (100x10(-8)M) induced a significant LH secretion after 2h of stimulus, reaching a maximum response 4h later. A rapid and remarkable LH release was induced by NEI (400x10(-8)M) 1h after stimulus, attaining its highest level at 2h. However, PRL, GH and FSH were not affected. NEI provoked ultrastructural changes in the gonadotrophs, which showed accumulations of LH-immunoreactive granules near the plasma membrane and exocytotic images, while the other populations exhibited no changes. Although NEI (10x10(-8)M), caused no action when used alone, its co-incubation with GnRH (1x10(-9)M), promoted a slight but significant increase in LH. These results demonstrate that NEI acts at the pituitary level through a direct action on gonadotrophs, as well as through interaction with GnRH.
Collapse
Affiliation(s)
- Ana Lucía De Paul
- Centro de Microscopía Electrónica, Universidad Nacional de Córdoba, Ciudad Universitaria, Argentina.
| | | | | | | | | | | | | |
Collapse
|
99
|
Poon SL, Hammond GT, Leung PCK. Epidermal growth factor-induced GnRH-II synthesis contributes to ovarian cancer cell invasion. Mol Endocrinol 2009; 23:1646-56. [PMID: 19608641 DOI: 10.1210/me.2009-0147] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH-II modulates ovarian cancer cells invasion and is expressed in normal ovary and ovarian epithelial cancer cells; however, the upstream regulator(s) of GnRH-II expression in these cells remains unclear. We now demonstrate that epidermal growth factor (EGF) increases GnRH-II mRNA levels in several human ovarian carcinoma cell lines and up-regulates GnRH-II promoter activity in OVCAR-3 cells in a dose-dependent manner, whereas an EGF receptor inhibitor (AG148) abolishes EGF-induced increases in GnRH-II promoter activity and GnRH-II mRNA levels. EGF increases the phosphorylation of cAMP-responsive element-binding protein (p-CREB) and its association with the coregulator, CCAAT/enhancer binding protein beta, whereas blocking the EGF-induced ERK1/2 phosphorylation with MAPK inhibitors (PD98059/U0126) markedly reduced these effects. Moreover, depletion of CREB using small interfering RNA attenuated EGF-induced GnRH-II promoter activity. Chromatin immunoprecipitation assays demonstrated that EGF induces p-CREB binding to a cAMP responsive-element within the GnRH-II promoter, likely in association with CCAAT/enhancer binding protein beta, and mutagenesis of this cAMP responsive-element prevented EGF-induced GnRH-II promoter activity in OVCAR-3 cells. Importantly, GnRH-II acts additively with EGF to promote invasion of OVCAR-3 and CaOV-3 cells, but not SKOV-3 cells that express low levels of GnRH receptor (GnRHR). Treatment with GnRHR small interfering RNA also partially inhibited the EGF-induced invasion of OVCAR-3 and CaOV-3 cells. Furthermore, EGF treatment transiently increases GnRHR levels in OVCAR-3 and CaOV-3, which likely accentuates the effects of increase GnRH-II production on cell invasion. These results provide evidence that EGF is an upstream regulator of the autocrine actions of GnRH-II on the invasive properties of ovarian cancer cells.
Collapse
Affiliation(s)
- Song Ling Poon
- Department of Obstetrics and Gynaecology, Child and Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5
| | | | | |
Collapse
|
100
|
Pearce WJ. Multifunctional angiogenic factors: add GnRH to the list. Focus on "Gonadotropin-releasing hormone-regulated chemokine expression in human placentation". Am J Physiol Cell Physiol 2009; 297:C4-5. [PMID: 19439527 DOI: 10.1152/ajpcell.00209.2009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|