1
|
Toufaily C, Fortin J, Alonso CA, Lapointe E, Zhou X, Santiago-Andres Y, Lin YF, Cui Y, Wang Y, Devost D, Roelfsema F, Steyn F, Hanyaloglu AC, Hébert TE, Fiordelisio T, Boerboom D, Bernard DJ. Addition of a carboxy terminal tail to the normally tailless gonadotropin-releasing hormone receptor impairs fertility in female mice. eLife 2021; 10:72937. [PMID: 34939930 PMCID: PMC8741216 DOI: 10.7554/elife.72937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/06/2021] [Indexed: 11/13/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is the primary neuropeptide controlling reproduction in vertebrates. GnRH stimulates follicle-stimulating hormone (FSH) and luteinizing hormone (LH) synthesis via a G-protein-coupled receptor, GnRHR, in the pituitary gland. In mammals, GnRHR lacks a C-terminal cytosolic tail (Ctail) and does not exhibit homologous desensitization. This might be an evolutionary adaptation that enables LH surge generation and ovulation. To test this idea, we fused the chicken GnRHR Ctail to the endogenous murine GnRHR in a transgenic model. The LH surge was blunted, but not blocked in these mice. In contrast, they showed reductions in FSH production, ovarian follicle development, and fertility. Addition of the Ctail altered the nature of agonist-induced calcium signaling required for normal FSH production. The loss of the GnRHR Ctail during mammalian evolution is unlikely to have conferred a selective advantage by enabling the LH surge. The adaptive significance of this specialization remains to be determined.
Collapse
Affiliation(s)
- Chirine Toufaily
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Jérôme Fortin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Carlos Ai Alonso
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Evelyne Lapointe
- Département de biomédecine vétérinaire, Universite de Montreal, Ste-Hyacinthe, Canada
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Yorgui Santiago-Andres
- Departamento de Ecología y Recursos Naturales, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yeu-Farn Lin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Yiming Cui
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Ying Wang
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Dominic Devost
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Ferdinand Roelfsema
- Department of Internal Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Frederik Steyn
- School of Biomedical Sciences, The University of Queensland, Brisbane, Australia
| | - Aylin C Hanyaloglu
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| | - Tatiana Fiordelisio
- 3epartamento de Ecología y Recursos Naturales, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Derek Boerboom
- Département de biomédecine vétérinaire, Universite de Montreal, Ste-Hyacinthe, Canada
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Canada
| |
Collapse
|
2
|
Newton CL, Anderson RC, Kreuchwig A, Krause G, Katz AA, Millar RP. Rescue of Function of Mutant Luteinising Hormone Receptors with Deficiencies in Cell Surface Expression, Hormone Binding, and Hormone Signalling. Neuroendocrinology 2021; 111:451-464. [PMID: 32316022 DOI: 10.1159/000508000] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/18/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION G protein-coupled receptor (GPCR) mutations are implicated in many diseases. Most inactivating mutations cause receptor misfolding and prevent trafficking to the plasma membrane. Pharmacological chaperones can "rescue" cell surface expression of such mutants, presumably by stabilising correct folding of the nascent protein. OBJECTIVE Here we examine the scope of intracellularly retained luteinising hormone receptor (LHR) mutants that can be "rescued" by the pharmacological chaperone LHR-Chap, and whether this allosteric agonist can also restore the function of mutant LHRs with deficiencies in hormone binding or hormone-induced signalling. METHODS Mutant LHRs were expressed in HEK 293-T cells. Cell surface expression/localisation, hormone binding, and hCG/LHR-Chap signalling were determined by ELISA, radioligand binding, and inositol phosphate accumulation assays, respectively. Molecular modelling predicted LHR-Chap interactions. RESULTS LHR-Chap increased cell surface expression of a subset of retained mutants located in transmembrane helices predicted to be stabilised by LHR-Chap binding. For 3 (T4613.47I, L5024.61P, and S6167.46Y) hCG-responsiveness was increased following treatment. LHRs with mutations in the hormone-binding site (C131ECDR and I152ECDT) or in the hinge region (E354HingeK) had good cell surface expression but poor response to hormone stimulation, yet were responsive to allosteric activation by LHR-Chap. CONCLUSIONS LHR-Chap, in addition to rescuing cell surface expression of intracellularly retained LHR mutants, can rescue function in mutant receptors with binding and signalling deficiencies that have normal cell surface expression. This demonstration of rescue of multiple elements of LHR dysfunction arising from inactivating mutations offers exceptional potential for treating patients with diseases arising from GPCR mutations in general.
Collapse
Affiliation(s)
- Claire Louise Newton
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa,
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa,
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom,
| | - Ross Calley Anderson
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Annika Kreuchwig
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Gerd Krause
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Arieh Anthony Katz
- UCT Receptor Biology Research Unit and SAMRC Gynaecology Cancer Research Centre, Department of Integrative Biomedical Sciences and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Robert Peter Millar
- Centre for Neuroendocrinology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- School of Medicine, University of St Andrews, St Andrews, United Kingdom
| |
Collapse
|
3
|
Huerta-Reyes M, Maya-Núñez G, Pérez-Solis MA, López-Muñoz E, Guillén N, Olivo-Marin JC, Aguilar-Rojas A. Treatment of Breast Cancer With Gonadotropin-Releasing Hormone Analogs. Front Oncol 2019; 9:943. [PMID: 31632902 PMCID: PMC6779786 DOI: 10.3389/fonc.2019.00943] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022] Open
Abstract
Although significant progress has been made in the implementation of new breast cancer treatments over the last three decades, this neoplasm annually continues to show high worldwide rates of morbidity and mortality. In consequence, the search for novel therapies with greater effectiveness and specificity has not come to a stop. Among the alternative therapeutic targets, the human gonadotropin-releasing hormone type I and type II (hGnRH-I and hGnRH–II, respectively) and its receptor, the human gonadotropin-releasing hormone receptor type I (hGnRHR-I), have shown to be powerful therapeutic targets to decrease the adverse effects of this disease. In the present review, we describe how the administration of GnRH analogs is able to reduce circulating concentrations of estrogen in premenopausal women through their action on the hypothalamus–pituitary–ovarian axis, consequently reducing the growth of breast tumors and disease recurrence. Also, it has been mentioned that, regardless of the suppression of synthesis and secretion of ovarian steroids, GnRH agonists exert direct anticancer action, such as the reduction of tumor growth and cell invasion. In addition, we discuss the effects on breast cancer of the hGnRH-I and hGnRH-II agonist and antagonist, non-peptide GnRH antagonists, and cytotoxic analogs of GnRH and their implication as novel adjuvant therapies as antitumor agents for reducing the adverse effects of breast cancer. In conclusion, we suggest that the hGnRH/hGnRHR system is a promising target for pharmaceutical development in the treatment of breast cancer, especially for the treatment of advanced states of this disease.
Collapse
Affiliation(s)
- Maira Huerta-Reyes
- Unidad de Investigación Médica en Enfermedades Nefrológicas, Centro Médico Nacional Siglo XXI (CMN-SXXI), Instituto Mexicano del Seguro Social (IMSS), Hospital de Especialidades, Mexico City, Mexico
| | - Guadalupe Maya-Núñez
- Unidad de Investigación Médica en Medicina Reproductiva, IMSS, Unidad Médica de Alta Especialidad No. 4, Mexico City, Mexico
| | - Marco Allán Pérez-Solis
- Unidad de Investigación Médica en Medicina Reproductiva, IMSS, Unidad Médica de Alta Especialidad No. 4, Mexico City, Mexico
| | - Eunice López-Muñoz
- Unidad de Investigación Médica en Medicina Reproductiva, IMSS, Unidad Médica de Alta Especialidad No. 4, Mexico City, Mexico
| | - Nancy Guillén
- Centre National de la Recherche Scientifique, CNRS-ERL9195, Paris, France
| | - Jean-Christophe Olivo-Marin
- Unité d'Analyse d'Images Biologiques, Institut Pasteur, Paris, France.,Centre National de la Recherche Scientifique, CNRS-UMR3691, Paris, France
| | - Arturo Aguilar-Rojas
- Unidad de Investigación Médica en Medicina Reproductiva, IMSS, Unidad Médica de Alta Especialidad No. 4, Mexico City, Mexico.,Unité d'Analyse d'Images Biologiques, Institut Pasteur, Paris, France
| |
Collapse
|
4
|
Drug screening for Pelizaeus-Merzbacher disease by quantifying the total levels and membrane localization of PLP1. Mol Genet Metab Rep 2019; 20:100474. [PMID: 31110947 PMCID: PMC6510973 DOI: 10.1016/j.ymgmr.2019.100474] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/28/2019] [Indexed: 01/01/2023] Open
Abstract
Background Pelizaeus-Merzbacher disease (PMD) is caused by point mutations or copy number changes in the proteolipid protein 1 gene (PLP1). PLP1 is exclusively localized in the myelin sheath of oligodendrocytes. Amino acid-substituted PLP1 protein is unable to fold properly and is subsequently degraded and/or restrictedly translated, resulting in a decrease in the PLP1 protein level and a failure to localize to the membrane. Furthermore, misfolded proteins increase the burden on the intracellular quality control system and trafficking, finally resulting in cell apoptosis. The objective of this study was to identify therapeutic chemicals for PMD by quantifying the total levels and membrane localization of PLP1. Method We established a cell line stably expressing PLP1A243V fused with green fluorescent protein in oligodendrocyte-derived MO3.13 cells. We screened a chemical library composed of drugs approved for central nervous system disorders that increased both the total intensity of PLP1A243V in the whole cell and the cell membrane localization. We analyzed the change in the endoplasmic reticulum (ER) stress and the gene expression of candidate chemicals using a micro-array analysis. Finally, we tested the in vivo effectiveness using myelin synthesis deficient (msd) mice with PlpA243V. Results and conclusion Piracetam significantly increased the PLP1A243V intensity and membrane localization and decreased the ER stress. It was also shown to reverse the gene expression changes induced by PLP1A243V in a micro-array analysis. However, in vivo treatment of piracetam did not improve the survival of msd mice (Plp1A243V).
Collapse
Key Words
- CNS, Central nervous systems
- Drug screening
- EGFP, Enhanced green fluorescent protein
- ER, Endoplasmic reticulum
- ER-associated degradation
- Gene expression
- IPA, Ingenuity pathways analysis
- IRE1 α, Inositol requiring enzyme 1 α
- Membrane protein
- Oligodendrocyte
- PLP1
- PLP1, Proteolipid protein 1
- PMD, Pelizaeus-Merzbacher disease
- UPR, Unfolded protein response
- XBP1, X-box binding protein 1
- msd, Myelin synthesis deficient
Collapse
|
5
|
Rima UK, Bari ASM, Hossain MZ, Khan MAH. Plasmid DNA vaccine coding eight repeats of gonadotrophin-releasing hormone induced atrophy of prostate in male mice. Prostate Int 2018; 6:151-156. [PMID: 30505818 PMCID: PMC6251954 DOI: 10.1016/j.prnil.2018.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 11/17/2017] [Accepted: 01/04/2018] [Indexed: 11/26/2022] Open
Abstract
Background Prostate hyperplasia and neoplasia are major illness of men and elderly dogs. Treatment of prostate cancer requires androgen deprivation surgery or therapy to prevent metastases and alleviate pain. Recently, six DNA vaccines have entered clinical trials against prostate cancer in humans with limited success. There is a need for new therapies that delay the establishment of malignancy and prolong survival. Materials and methods A plasmid DNA vaccine coding for eight gonadotrophin-releasing hormone (GnRH-I) interspersed in eight T-helper epitopes was used. Sexually mature male mice were immunized with the vaccine in hemagglutinating virus of Japanese envelope vector and boosted in nonionized surfactant vesicles in study weeks 0, 3, 6, 9, and 12. Plasma anti-GnRH-I antibody response, serum testosterone concentration, and effect on prostate were evaluated. Results Results of an indirect enzyme linked immunosorbent assay (ELISA) showed anti-GnRH-I antibody response (OD value) detected in the study week 3 (0.613 ± 0.179) with a highest response in the week 12 (1.205 ± 0.219). Serum testosterone concentration (ng/ml) in vaccinated mice was significantly reduced (P > 0.000, 0.761 ± 0.531) in the study week 24 in contrast to control serum (7.583 ± 1.251). Group average gross combined weight of prostate and seminal vesicles of vaccinated mice was significantly (P < 0.000) reduced in the study week 24 (319.75 ± 89.19 mg) in contrast to control weight (563.25 ± 108.60 mg). Sections of prostate stained with Goldner's trichrome showed profuse pink color secretion in control tubules, which however was absent in the vaccinated prostate. The lining epithelium of the vaccinated prostate was atrophied and did not enfold in its lumen. Conclusions Immunization strategy designed with the plasmid DNA vaccine in hemagglutinating virus of Japanese envelope and nonionized surfactant vesicles can be the genetic immunization platform. This vaccine bears potentials in terms of reducing serum testosterone concentration and induction of atrophy of prostate. Targeted ablation of native GnRH-I by genetic immunization could offer leverage to vaccinologists, seeking therapeutic target to control and prevent malignancy of prostate.
Collapse
Affiliation(s)
- Umme K Rima
- Department of Medicine, Surgery and Obstetrics, Faculty of Veterinary & Animal Science, Hajee Mohammad Danesh Science & Technology University, Dinajpur, Bangladesh
| | - Abu S M Bari
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Mohammad Z Hossain
- Department of Livestock Services, Ministry of Livestock and Fisheries, Bangladesh
| | - Mohammad A H Khan
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, Bangladesh
| |
Collapse
|
6
|
Oláh G, Dobos N, Vámosi G, Szabó Z, Sipos É, Fodor K, Harda K, Schally AV, Halmos G. Experimental therapy of doxorubicin resistant human uveal melanoma with targeted cytotoxic luteinizing hormone-releasing hormone analog (AN-152). Eur J Pharm Sci 2018; 123:371-376. [PMID: 30076951 DOI: 10.1016/j.ejps.2018.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Cytotoxic analogs of LHRH (luteinizing hormone-releasing hormone) can be successfully used for the treatment of hormone-dependent cancers such as prostatic, ovarian, endometrial, but our knowledge about their effect on hormone-independent cancers such as human uveal melanoma (UM) is limited. Previously, we have demonstrated that 46% of UM express full-length LHRH receptors. This finding has led us to further examine the mechanism of action of LHRH receptor based targeted therapies in this malignancy. AIMS In the present study we investigated the cellular uptake of doxorubicin (DOX) and cytotoxic LHRH analog AN-152 (AEZS-108, zoptarelin doxorubicin) on human UM cell lines (OCM3) and its DOX resistant form OCM3DOX320 by confocal laser scanning microscopy. The LHRH receptor expression was characterized by RT-PCR and immunocytochemistry. RESULTS We were able to establish a new, stable and DOX resistant human UM cell line OCM3DOX320. Our results demonstrated the expression of splice variants and isoforms of receptor for LHRH in OCM3 UM cell line and its doxorubicin resistant form OCM3DOX320. It has been revealed by MTT assay that AN-152 inhibited cell proliferation in a dose dependent manner in OCM3DOX320 cells. Furthermore, receptor-mediated uptake of AN-152 was demonstrated using confocal laser scanning microscopy in both cell line. CONCLUSIONS Our results suggest that the antiproliferative effect of AN-152 can be detected even if only LHRH receptor isoforms are expressed. Our study also demonstrates the LHRH receptor-mediated uptake of AN-152 in DOX resistant OCM3DOX320 cells. Our experiments provide new insights into a potential targeted therapy of UM and give further details about the accumulation of AN-152 in hormone-independent DOX-resistant cells expressing splice variants of the LHRH receptors.
Collapse
Affiliation(s)
- Gábor Oláh
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Dobos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - György Vámosi
- Department of Biophysics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Szabó
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Éva Sipos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klára Fodor
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Kristóf Harda
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA; Department of Pathology, University of Miami, Miami, FL, USA; Department of Medicine, Division of Hematology-Oncology, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA; Department of Medicine, Division of Endocrinology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gábor Halmos
- Department of Biopharmacy, Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary; Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA.
| |
Collapse
|
7
|
Sipos E, Dobos N, Rozsa D, Fodor K, Olah G, Szabo Z, Szekvolgyi L, Schally AV, Halmos G. Characterization of luteinizing hormone-releasing hormone receptor type I (LH-RH-I) as a potential molecular target in OCM-1 and OCM-3 human uveal melanoma cell lines. Onco Targets Ther 2018; 11:933-941. [PMID: 29503568 PMCID: PMC5826244 DOI: 10.2147/ott.s148174] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Introduction Uveal melanoma (UM) is the most common primary intraocular malignancy with very poor prognosis. Conventional chemotherapy only rarely prolongs the survival, therefore patients require novel treatment modalities. The discovery of specific receptors for hypothalamic hormones on cancer cells has led to the development of radiolabeled and cytotoxic hormone analogs. Materials and methods In the present study, our aim was to investigate the expression of mRNA for receptors of luteinizing hormone-releasing hormone type I (LH-RH-I) and LH-RH ligand in OCM-1 and OCM-3 human uveal melanoma cell lines. The presence and binding characteristics of LH-RH-I receptor protein was further studied by Western blot, immunocytochemistry and ligand competition assay. The expression of mRNA and protein for LH-RH-I receptors has been also studied using tumor samples originating from nude mice xenografted with OCM-1 or OCM-3 cells. Results The mRNA for LH-RH-I receptor has been detected in OCM-1 and OCM-3 cell lines and was found markedly higher in OCM-3 cells. The mRNA for LH-RH-I receptors was also observed in both UM xenograft models in vivo with higher levels in OCM-3. The presence of LH-RH-I receptor protein was found in both cell lines in vitro by immunocytochemistry and Western blot, and also in tumor tissue samples grown in nude mice by Western blot. Both human uveal melanoma models investigated showed specific high affinity receptors for LH-RH-I using ligand competition assay. The mRNA for LH-RH ligand has also been detected in OCM-1 and OCM-3 cell lines and cancer tissues. Conclusion The demonstration of the expression of LH-RH-I receptors in OCM-1 and OCM-3 human UM cell lines suggests that they could serve as potential molecular target for therapy. Our findings support the development of new therapeutic approaches based on cytotoxic LH-RH analogs or modern powerful antagonistic analogs of LH-RH targeting LH-RH-I receptors in UM.
Collapse
Affiliation(s)
- Eva Sipos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Nikoletta Dobos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - David Rozsa
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Klara Fodor
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Gabor Olah
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Zsuzsanna Szabo
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Lorant Szekvolgyi
- MTA-DE Momentum, Genome Architecture and Recombination Research Group, Debrecen, Hungary.,Research Centre for Molecular Medicine; Department of Biochemistry and Molecular Biology, Debrecen, Hungary
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA.,Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA.,Department of Medicine, Divisions of Hematology-Oncology and Endocrinology, Miller School of Medicine, University of Miami, Miami, FL, USA.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Gabor Halmos
- Department of Biopharmacy, School of Pharmacy, University of Debrecen, Debrecen, Hungary.,Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL, USA
| |
Collapse
|
8
|
Aguilar-Rojas A, Maya-Núñez G, Huerta-Reyes M, Pérez-Solis MA, Silva-García R, Guillén N, Olivo-Marin JC. Activation of human gonadotropin-releasing hormone receptor promotes down regulation of ARHGAP18 and regulates the cell invasion of MDA-MB-231 cells. Mol Cell Endocrinol 2018; 460:94-103. [PMID: 28709956 DOI: 10.1016/j.mce.2017.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/29/2017] [Accepted: 07/10/2017] [Indexed: 02/08/2023]
Abstract
The Gonadotropin-Releasing Hormone Receptor (GnRHR) is expressed mainly in the gonadotrope membrane of the adenohypophysis and its natural ligand, the Gonadotropin-Releasing Hormone (GnRH), is produced in anterior hypothalamus. Furthermore, both molecules are also present in the membrane of cells derived from other reproductive tissues such as the breast, endometrium, ovary, and prostate, as well as in tumors derived from these tissues. The functions of GnRH receptor and its hormone in malignant cells have been related with the decrease of proliferation and the invasiveness of those tumors however, little is known about the molecules associated with the signaling pathways regulated by both molecules in malignant cells. To further analyze the potential mechanisms employed by the GnRHR/GnRH system to reduce the tumorigenesis of the highly invasive breast cancer cell line MDA-MB-231, we performed microarrays experiments to evaluated changes in genes expression and validate these modifications by functional assays. We show that activation of human GnRHR is able to diminish the expression and therefore functions of the Rho GTPase-Activating Protein 18 (ARHGAP18). Decrease of this GAP following GnRHR activation, correlates to the higher of cell adhesion and also with reduction of tumor cell invasion, supporting the notion that GnRHR triggers intracellular signaling pathways that acts through ARHGAP18. On the contrary, although a decline of cellular proliferation was observed during GnRHR activation in MDA-MB-231, this was independent of ARHGAP18 showing the complex system in which is involved the signaling pathways regulated by the GnRHR/GnRH system.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico; Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, F-75015 Paris, France; Centre National de la Recherche Scientifique, CNRS UMR3691, 25 Rue du Dr Roux, F-75015 Paris, France.
| | - Guadalupe Maya-Núñez
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico
| | - Maira Huerta-Reyes
- IMSS, Unidad de Investigación Médica en Farmacología, Hospital de Especialidades, Centro Médico Nacional Siglo XXI (CMN-SXXI), Ciudad de México, Mexico
| | - Marco Allán Pérez-Solis
- Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Medicina Reproductiva, UMAE No. 4, Ciudad de México, Mexico
| | - Raúl Silva-García
- IMSS, Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, CMN-SXXI, Ciudad de México, Mexico
| | - Nancy Guillén
- Centre National de la Recherche Scientifique, CNRS-ERL9195, 25 Rue du Dr Roux, F-75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, F-75015 Paris, France; Centre National de la Recherche Scientifique, CNRS UMR3691, 25 Rue du Dr Roux, F-75015 Paris, France
| |
Collapse
|
9
|
Abstract
The gonadotropin receptors (luteinising hormone receptor; LHR and follicle-stimulating hormone receptor; FSHR) are G protein-coupled receptors (GPCRs) that play an important role in the endocrine control of reproduction. Thus genetic mutations that cause impaired function of these receptors have been implicated in a number of reproductive disorders. Disease-causing genetic mutations in GPCRs frequently result in intracellular retention and degradation of the nascent protein through misfolding and subsequent recognition by cellular quality control machinery. The discovery and development of novel compounds termed pharmacological chaperones (pharmacoperones) that can stabilise misfolded receptors and restore trafficking and plasma membrane expression are therefore of great interest clinically, and promising in vitro data describing the pharmacoperone rescue of a number of intracellularly retained mutant GPCRs has provided a platform for taking these compounds into in vivo trials. Thienopyrimidine small molecule allosteric gonadotropin receptor agonists (Org 42599 and Org 41841) have been demonstrated to have pharmacoperone activity. These compounds can rescue cell surface expression and in many cases, hormone responsiveness, of a range of retained mutant gonadotropin receptors. Should gonadotropin receptor selectivity of these compounds be improved, they could offer therapeutic benefit to subsets of patients suffering from reproductive disorders attributed to defective gonadotropin receptor trafficking.
Collapse
Affiliation(s)
- Claire L Newton
- Centre for Neuroendocrinology and Department of Immunology, Faculty of Health Sciences, University of Pretoria, PO Box 2034, Pretoria, 0001, South Africa.
| | - Ross C Anderson
- Centre for Neuroendocrinology and Department of Immunology, Faculty of Health Sciences, University of Pretoria, PO Box 2034, Pretoria, 0001, South Africa
| |
Collapse
|
10
|
Busby ER, Sherwood NM. Gonadotropin-releasing hormone receptor (Gnrhr) gene knock out: Normal growth and development of sensory, motor and spatial orientation behavior but altered metabolism in neonatal and prepubertal mice. PLoS One 2017; 12:e0174452. [PMID: 28346489 PMCID: PMC5367835 DOI: 10.1371/journal.pone.0174452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 03/09/2017] [Indexed: 11/27/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is important in the control of reproduction, but its actions in non-reproductive processes are less well known. In this study we examined the effect of disrupting the GnRH receptor in mice to determine if growth, metabolism or behaviors that are not associated with reproduction were affected. To minimize the effects of other hormones such as FSH, LH and sex steroids, the neonatal-prepubertal period of 2 to 28 days of age was selected. The study shows that regardless of sex or phenotype in the Gnrhr gene knockout line, there was no significant difference in the daily development of motor control, sensory detection or spatial orientation among the wildtype, heterozygous or null mice. This included a series of behavioral tests for touch, vision, hearing, spatial orientation, locomotory behavior and muscle strength. Neither the daily body weight nor the final weight on day 28 of the kidney, liver and thymus relative to body weight varied significantly in any group. However by day 28, metabolic changes in the GnRH null females compared with wildtype females showed a significant reduction in inguinal fat pad weight normalized to body weight; this was accompanied by an increase in glucose compared with wildtype females shown by Student-Newman-Keuls Multiple Comparison test and Student's unpaired t tests. Our studies show that the GnRH-GnRHR system is not essential for growth or motor/sensory/orientation behavior during the first month of life prior to puberty onset. The lack of the GnRH-GnRHR axis, however, did affect females resulting in reduced subcutaneous inguinal fat pad weight and increased glucose with possible insulin resistance; the loss of the normal rise of estradiol at postnatal days 15-28 may account for the altered metabolism in the prepubertal female pups.
Collapse
Affiliation(s)
- Ellen R. Busby
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | |
Collapse
|
11
|
Pope GR, Tilve S, McArdle CA, Lolait SJ, O'Carroll AM. Agonist-induced internalization and desensitization of the apelin receptor. Mol Cell Endocrinol 2016; 437:108-119. [PMID: 27492965 PMCID: PMC5062952 DOI: 10.1016/j.mce.2016.07.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/30/2016] [Accepted: 07/30/2016] [Indexed: 12/14/2022]
Abstract
Apelin acts via the G protein-coupled apelin receptor (APJ) to mediate effects on cardiovascular and fluid homeostasis. G protein-coupled receptor (GPCR) trafficking has an important role in the regulation of receptor signalling pathways and cellular functions, however in the case of APJ the mechanisms and proteins involved in apelin-induced trafficking are not well understood. We generated a stable HEK-293 cell line expressing N-terminus HA-tagged mouse (m) APJ, and used a semi-automated imaging protocol to quantitate APJ trafficking and ERK1/2 activation following stimulation with [Pyr1]apelin-13. The mechanisms of [Pyr1]apelin-13-induced internalization and desensitization were explored using dominant-negative mutant (DNM) cDNA constructs of G protein-coupled receptor kinase 2 (GRK2), β-arrestin1, EPS15 and dynamin. The di-phosphorylated ERK1/2 (ppERK1/2) response to [Pyr1]apelin-13 desensitized during sustained stimulation, due to upstream APJ-specific adaptive changes. Furthermore, [Pyr1]apelin-13 stimulation caused internalization of mAPJ via clathrin coated vesicles (CCVs) and also caused a rapid reduction in cell surface and whole cell HA-mAPJ. Our data suggest that upon continuous agonist exposure GRK2-mediated phosphorylation targets APJ to CCVs that are internalized from the cell surface in a β-arrestin1-independent, EPS15- and dynamin-dependent manner. Internalization does not appear to contribute to the desensitization of APJ-mediated ppERK1/2 activation in these cells.
Collapse
Affiliation(s)
- George R Pope
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Sharada Tilve
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Craig A McArdle
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Stephen J Lolait
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| | - Anne-Marie O'Carroll
- Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK.
| |
Collapse
|
12
|
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: 1.8] [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
|
13
|
Abstract
Reproductive hormones affect all stages of life from gamete production, fertilization, fetal development and parturition, neonatal development and puberty through to adulthood and senescence. The reproductive hormone cascade has, therefore, been the target for the development of numerous drugs that modulate its activity at many levels. As the central regulator of the cascade, gonadotropin-releasing hormone (GnRH) agonists and antagonists have found extensive applications in treating a wide range of hormone-dependent diseases, such as precocious puberty, prostate cancer, benign prostatic hyperplasia, endometriosis and uterine fibroids, as well as being an essential component of in vitro fertilization protocols. The neuroendocrine peptides that regulate GnRH neurons, kisspeptin and neurokinin B, have also been identified as therapeutic targets, and novel agonists and antagonists are being developed as modulators of the cascade upstream of GnRH. Here, we review the development and applications of analogues of the major neuroendocrine peptide regulators of the reproductive hormone cascade: GnRH, kisspeptin and neurokinin B.
Collapse
Affiliation(s)
- Robert P Millar
- Mammal Research Institute, Department of Zoology and Entomology, Faculty of Natural and Agricultural Sciences, University of Pretoria, South Africa.
| | | |
Collapse
|
14
|
Tello JA, Kohout T, Pineda R, Maki RA, Scott Struthers R, Millar RP. Reproductive physiology of a humanized GnRH receptor mouse model: application in evaluation of human-specific analogs. Am J Physiol Endocrinol Metab 2013; 305:E67-77. [PMID: 23632635 DOI: 10.1152/ajpendo.00624.2012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The human GnRH receptor (GNRHR1) has a specific set of properties with physiological and pharmacological influences not appropriately modeled in laboratory animals or cell-based systems. To address this deficiency, we have generated human GNRHR1 knock-in mice and described their reproductive phenotype. Measurement of pituitary GNRHR1 transcripts from homozygous human GNRHR1 knock-in (ki/ki) mice revealed a severe reduction (7- to 8-fold) compared with the mouse Gnrhr1 in wild-type mice. ¹²⁵I-GnRH binding assays on pituitary membrane fractions corroborated reduced human GNRHR1 protein expression in ki/ki mice, as occurs with transfection of human GNRHR1 in cell lines. Female homozygous knock-in mice displayed normal pubertal onset, indicating that a large reduction in GNRHR1 expression is sufficient for this process. However, ki/ki females exhibited periods of prolonged estrous and/or metestrous and reduced fertility. No impairment was found in reproductive maturity or adult fertility in male ki/ki mice. Interestingly, the serum LH response to GnRH challenge was reduced in both knock-in males and females, indicating a reduced GNRHR1 signaling capacity. Small molecules targeting human GPCRs usually have poor activities at homologous rodent receptors, thus limiting their use in preclinical development. Therefore, we tested a human-specific GnRH1 antagonist, NBI-42902, in our mouse model and demonstrated abrogation of a GnRH1-induced serum LH rise in ki/ki mice and an absence of effect in littermates expressing the wild-type murine receptor. This novel model provides the opportunity to study the human receptor in vivo and for screening the activity of human-specific GnRH analogs.
Collapse
Affiliation(s)
- Javier A Tello
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | | | | |
Collapse
|
15
|
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: 0.4] [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
|
16
|
Aguilar-Rojas A, Huerta-Reyes M, Maya-Núñez G, Arechavaleta-Velásco F, Conn PM, Ulloa-Aguirre A, Valdés J. Gonadotropin-releasing hormone receptor activates GTPase RhoA and inhibits cell invasion in the breast cancer cell line MDA-MB-231. BMC Cancer 2012; 12:550. [PMID: 23176180 PMCID: PMC3518142 DOI: 10.1186/1471-2407-12-550] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 10/25/2012] [Indexed: 11/17/2022] Open
Abstract
Background Gonadotropin-releasing hormone (GnRH) and its receptor (GnRHR) are both expressed by a number of malignant tumors, including those of the breast. In the latter, both behave as potent inhibitors of invasion. Nevertheless, the signaling pathways whereby the activated GnRH/GnRHR system exerts this effect have not been clearly established. In this study, we provide experimental evidence that describes components of the mechanism(s) whereby GnRH inhibits breast cancer cell invasion. Methods Actin polymerization and substrate adhesion was measured in the highly invasive cell line, MDA-MB-231 transiently expressing the wild-type or mutant DesK191 GnRHR by fluorometry, flow cytometric analysis, and confocal microscopy, in the absence or presence of GnRH agonist. The effect of RhoA-GTP on stress fiber formation and focal adhesion assembly was measured in MDA-MB-231 cells co-expressing the GnRHRs and the GAP domain of human p190Rho GAP-A or the dominant negative mutant GAP-Y1284D. Cell invasion was determined by the transwell migration assay. Results Agonist-stimulated activation of the wild-type GnRHR and the highly plasma membrane expressed mutant GnRHR-DesK191 transiently transfected to MDA-MB-231 cells, favored F-actin polymerization and substrate adhesion. Confocal imaging allowed detection of an association between F-actin levels and the increase in stress fibers promoted by exposure to GnRH. Pull-down assays showed that the effects observed on actin cytoskeleton resulted from GnRH-stimulated activation of RhoA GTPase. Activation of this small G protein favored the marked increase in both cell adhesion to Collagen-I and number of focal adhesion complexes leading to inhibition of the invasion capacity of MDA-MB-231 cells as disclosed by assays in Transwell Chambers. Conclusions We here show that GnRH inhibits invasion of highly invasive breast cancer-derived MDA-MB-231 cells. This effect is mediated through an increase in substrate adhesion promoted by activation of RhoA GTPase and formation of stress fibers and focal adhesions. These observations offer new insights into the molecular mechanisms whereby activation of overexpressed GnRHRs affects cell invasion potential of this malignant cell line, and provide opportunities for designing mechanism-based adjuvant therapies for breast cancer.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Centro de Investigación Biomédica del Sur (CIBIS), Instituto Mexicano del Seguro Social (IMSS), Argentina No, 1, Col, Centro, 62790, Xochitepec, Morelos, Mexico.
| | | | | | | | | | | | | |
Collapse
|
17
|
Hazell GG, Hindmarch CC, Pope GR, Roper JA, Lightman SL, Murphy D, O’Carroll AM, Lolait SJ. G protein-coupled receptors in the hypothalamic paraventricular and supraoptic nuclei--serpentine gateways to neuroendocrine homeostasis. Front Neuroendocrinol 2012; 33:45-66. [PMID: 21802439 PMCID: PMC3336209 DOI: 10.1016/j.yfrne.2011.07.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 06/24/2011] [Accepted: 07/06/2011] [Indexed: 12/31/2022]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of transmembrane receptors in the mammalian genome. They are activated by a multitude of different ligands that elicit rapid intracellular responses to regulate cell function. Unsurprisingly, a large proportion of therapeutic agents target these receptors. The paraventricular nucleus (PVN) and supraoptic nucleus (SON) of the hypothalamus are important mediators in homeostatic control. Many modulators of PVN/SON activity, including neurotransmitters and hormones act via GPCRs--in fact over 100 non-chemosensory GPCRs have been detected in either the PVN or SON. This review provides a comprehensive summary of the expression of GPCRs within the PVN/SON, including data from recent transcriptomic studies that potentially expand the repertoire of GPCRs that may have functional roles in these hypothalamic nuclei. We also present some aspects of the regulation and known roles of GPCRs in PVN/SON, which are likely complemented by the activity of 'orphan' GPCRs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Stephen J. Lolait
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, Dorothy Hodgkin Building, School of Clinical Sciences, University of Bristol, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
18
|
Lirón JP, Prando A, Ripoli MV, Rogberg-Muñoz A, Posik DM, Baldo A, Peral-García P, Giovambattista G. Characterization and validation of bovine gonadotripin releasing hormone receptor (GNRHR) polymorphisms. Res Vet Sci 2011; 91:391-6. [PMID: 21030057 DOI: 10.1016/j.rvsc.2010.09.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 09/21/2010] [Accepted: 09/26/2010] [Indexed: 11/28/2022]
Abstract
Gonadotropin releasing hormone and its receptor (GNRHR) play a critical role in sexual differentiation and reproduction. Available evidence shows a strong genetic component in the timing of puberty. In bovines, there are significant differences within and among beef breeds in the time when bulls reach puberty. Despite its economic importance, there are not many SNPs or genetic markers associated with this characteristic. The aims of the study were to identify DNA polymorphism in the bovine GNRHR by re-sequencing analysis, determine haplotype phases, and perform a population study in a selected tag SNP in six breeds. Eight SNPs were detected, including: one in the Upstream Regulatory Region (URR), five in the coding regions, and two in non-coding regions. This polymorphism level corresponds to one variant every 249.4bp and a global nucleotide diversity of 0.385. Two haplogroups comprising nine haplotypes and two linkage blocks were detected. Despite 5 tag SNPs were required to capture all variability, just one SNP allowed to define both haplogroups, and only two SNPs were needed to differentiate the most common haplotypes. An additional taq SNP was necessary to identify both URR variants. Allele-frequency analysis of a selected taq SNP among breeds showed a geographical cline. European Bos taurus breeds had lower frequencies of the C allele than B. indicus type cattle, while Creole cattle and Wagyu breeds had intermediate frequency. There was a significant correlation between frequency profile and timing of puberty among the studied breeds, which seems to suggest that genetic variation within bovine GNRHR gene could explain at least part of the reported variability.
Collapse
Affiliation(s)
- J P Lirón
- Instituto de Genética Veterinaria (IGEVET), CCT La Plata - CONICET-Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, Calle 60 y 118 s/n, CC 296, 1900 La Plata, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Chevrier L, Guimiot F, de Roux N. GnRH receptor mutations in isolated gonadotropic deficiency. Mol Cell Endocrinol 2011; 346:21-8. [PMID: 21645587 DOI: 10.1016/j.mce.2011.04.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 04/02/2011] [Indexed: 10/18/2022]
Abstract
GnRH and its receptor GnRHR are key regulators of the hypothalamo-pituitary axis. They modulate the secretion of LH and FSH gonadotropins and therefore, the development and maturation of gonads in fetal life as well as after birth. Congenital functional defect of this axis results in isolated hypogonadotropic hypogonadism (IHH). Several natural mutations causing IHH without anosmia have now been identified in GnRHR or GnRH genes. These mutations inactivate GnRHR or its ligand function and cause highly variable phenotypes, ranging from partial to complete gonadotropic deficiencies. The present review describes the published natural GnRHR mutations and tries to correlate them with the corresponding phenotypes according to the different steps of the GnRH system development.
Collapse
Affiliation(s)
- L Chevrier
- INSERM U676, Avenir Team: Genetic and Physiology of Puberty Onset, Robert Debre Hospital, 48 Boulevard Serurier, 75019 Paris, France
| | | | | |
Collapse
|
20
|
Rescue of expression and signaling of human luteinizing hormone G protein-coupled receptor mutants with an allosterically binding small-molecule agonist. Proc Natl Acad Sci U S A 2011; 108:7172-6. [PMID: 21482767 DOI: 10.1073/pnas.1015723108] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Naturally occurring mutations of G protein-coupled receptors (GPCRs) causing misfolding and failure to traffic to the cell surface can result in disease states. Some small-molecule orthosteric ligands can rescue such misfolded receptors, presumably by facilitating their correct folding and shuttling to the plasma membrane. Here we show that a cell-permeant, allosterically binding small-molecule agonist (Org 42599) rescues the folding and cell surface expression, and therefore target cell signaling, of mutant human luteinizing hormone (LH) receptors (A593P and S616Y) that cause Leydig cell hypoplasia in man. Both mutant receptors were retained in the cytoplasm whereas WT receptor localized at the cell membrane, and binding of LH to cells expressing the mutant receptors was markedly lower than to those expressing the WT receptor. Incubation with Org 42599 increased mutant receptor expression, cell surface localization, and the proportion of mutant receptor in the mature glycosylated form. Importantly, although LH stimulated little (S616Y) or no (A593P) activation of cells expressing mutant receptors, incubation of cells with Org 42599 facilitated rescue of expression and stimulation by the native ligand, LH. Although Org 42599 could activate these receptors, it could not displace (125)I-labeled human LH binding to the WT receptor, indicating that it acts in an allosteric manner. Here we demonstrate a small-molecule GPCR allosteric agonist that functionally rescues intracellularly retained mutant LH receptors by facilitating their cell surface expression. This approach may have application for treatment of infertile patients bearing such mutations and, more broadly, for other misfolded GPCR mutants resulting in human pathologic processes.
Collapse
|
21
|
Armstrong S, Caunt C, Finch A, McArdle C. Using automated imaging to interrogate gonadotrophin-releasing hormone receptor trafficking and function. Mol Cell Endocrinol 2011; 331:194-204. [PMID: 20688134 PMCID: PMC3021717 DOI: 10.1016/j.mce.2010.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 05/07/2010] [Accepted: 07/13/2010] [Indexed: 01/03/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH) acts via seven transmembrane receptors on gonadotrophs to stimulate gonadotrophin synthesis and secretion, and thereby mediates central control of reproduction. Type I mammalian GnRHR are unique, in that they lack C-terminal tails. This is thought to underlie their resistance to rapid homologous desensitisation as well as their slow rate of internalisation and inability to provoke G-protein-independent (arrestin-mediated) signalling. More recently it has been discovered that the vast majority of human GnRHR are actually intracellular, in spite of the fact that they are activated at the cell surface by a membrane impermeant peptide hormone. This apparently reflects inefficient exit from the endoplasmic reticulum and again, the absence of the C-tail likely contributes to their intracellular localisation. This review is intended to cover some of these novel aspects of GnRHR biology, focusing on ways that we have used automated fluorescence microscopy (high content imaging) to explore GnRHR localisation and trafficking as well as spatial and temporal aspects of GnRH signalling via the Ca(2+)/calmodulin/calcineurin/NFAT and Raf/MEK/ERK pathways.
Collapse
Affiliation(s)
- S.P. Armstrong
- University of Bristol, School of Clinical Sciences, Labs. for Integrative Neuroscience and Endocrinology, 1 Whitson Street, Bristol BS1 3NY, UK
| | - C.J. Caunt
- Department of Biology & Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - A.R. Finch
- University of Bristol, School of Clinical Sciences, Labs. for Integrative Neuroscience and Endocrinology, 1 Whitson Street, Bristol BS1 3NY, UK
| | - C.A. McArdle
- University of Bristol, School of Clinical Sciences, Labs. for Integrative Neuroscience and Endocrinology, 1 Whitson Street, Bristol BS1 3NY, UK
- Corresponding author.
| |
Collapse
|
22
|
Re M, Pampillo M, Savard M, Dubuc C, McArdle CA, Millar RP, Conn PM, Gobeil F, Bhattacharya M, Babwah AV. The human gonadotropin releasing hormone type I receptor is a functional intracellular GPCR expressed on the nuclear membrane. PLoS One 2010; 5:e11489. [PMID: 20628612 PMCID: PMC2900216 DOI: 10.1371/journal.pone.0011489] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Accepted: 06/11/2010] [Indexed: 12/02/2022] Open
Abstract
The mammalian type I gonadotropin releasing hormone receptor (GnRH-R) is a structurally unique G protein-coupled receptor (GPCR) that lacks cytoplasmic tail sequences and displays inefficient plasma membrane expression (PME). Compared to its murine counterparts, the primate type I receptor is inefficiently folded and retained in the endoplasmic reticulum (ER) leading to a further reduction in PME. The decrease in PME and concomitant increase in intracellular localization of the mammalian GnRH-RI led us to characterize the spatial distribution of the human and mouse GnRH receptors in two human cell lines, HEK 293 and HTR-8/SVneo. In both human cell lines we found the receptors were expressed in the cytoplasm and were associated with the ER and nuclear membrane. A molecular analysis of the receptor protein sequence led us to identify a putative monopartite nuclear localization sequence (NLS) in the first intracellular loop of GnRH-RI. Surprisingly, however, neither the deletion of the NLS nor the addition of the Xenopus GnRH-R cytoplasmic tail sequences to the human receptor altered its spatial distribution. Finally, we demonstrate that GnRH treatment of nuclei isolated from HEK 293 cells expressing exogenous GnRH-RI triggers a significant increase in the acetylation and phosphorylation of histone H3, thereby revealing that the nuclear-localized receptor is functional. Based on our findings, we conclude that the mammalian GnRH-RI is an intracellular GPCR that is expressed on the nuclear membrane. This major and novel discovery causes us to reassess the signaling potential of this physiologically and clinically important receptor.
Collapse
Affiliation(s)
- Michelle Re
- The Children's Health Research Institute, London, Canada
- Lawson Health Research Institute, London, Canada
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Canada
| | - Macarena Pampillo
- The Children's Health Research Institute, London, Canada
- Lawson Health Research Institute, London, Canada
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Canada
| | - Martin Savard
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Canada
| | - Céléna Dubuc
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Canada
| | - Craig A. McArdle
- Laboratories for Integrated Neuroscience and Endocrinology, Department of Clinical Sciences at South Bristol, University of Bristol, Bristol, United Kingdom
| | - Robert P. Millar
- MRC Human Reproductive Sciences Unit, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - P. Michael Conn
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, United States of America
| | - Fernand Gobeil
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, Canada
| | - Moshmi Bhattacharya
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Canada
| | - Andy V. Babwah
- The Children's Health Research Institute, London, Canada
- Lawson Health Research Institute, London, Canada
- Department of Obstetrics and Gynaecology, The University of Western Ontario, London, Canada
- Department of Physiology and Pharmacology, The University of Western Ontario, London, Canada
| |
Collapse
|
23
|
Armstrong SP, Caunt CJ, Fowkes RC, Tsaneva-Atanasova K, McArdle CA. Pulsatile and sustained gonadotropin-releasing hormone (GnRH) receptor signaling: does the Ca2+/NFAT signaling pathway decode GnRH pulse frequency? J Biol Chem 2010; 284:35746-57. [PMID: 19858197 PMCID: PMC2791005 DOI: 10.1074/jbc.m109.063917] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) acts via 7 transmembrane region receptors on gonadotrophs to stimulate synthesis and secretion of the luteinizing hormone and follicle-stimulating hormone. It is secreted in pulses, and its effects depend on pulse frequency, but decoding mechanisms are unknown. Here we have used (nuclear factor of activated T-cells 2 (NFAT2)-emerald fluorescent protein) to monitor GnRH signaling. Increasing [Ca2+]i causes calmodulin/calcineurin-dependent nuclear NFAT translocation, a response involving proteins (calmodulins and NFATs) that decode frequency in other systems. Using live cell imaging, pulsatile GnRH caused dose- and frequency-dependent increases in nuclear NFAT2-emerald fluorescent protein, and at low frequency, translocation simply tracked GnRH exposure (albeit with slower kinetics). At high frequency (30-min intervals), failure to return to basal conditions before repeat stimulation caused integrative tracking, illustrating how the relative dynamics of up- and downstream signals can increase efficiency of GnRH action. Mathematical modeling predicted desensitization of GnRH effects on [Ca2+]i and that desensitization would increase with dose, frequency, and receptor number, but no such desensitization was seen in HeLa and/or LβT2 cells possibly because pulsatile GnRH did not reduce receptor expression (measured by immunofluorescence). GnRH also caused dose- and frequency-dependent activation of αGSU, luteinizing hormone β, and follicle-stimulating hormone β luciferase reporters, effects that were blocked by calcineurin inhibition. Pulsatile GnRH also activated an NFAT-responsive luciferase reporter, but this response was directly related to cumulative pulse duration. This together with the lack of desensitization of translocation responses suggests that NFAT may mediate GnRH action but is not a genuine decoder of GnRH pulse frequency.
Collapse
Affiliation(s)
- Stephen P Armstrong
- Laboratories for Integrative Neuroscience and Endocrinology, Department of Clinical Science at South Bristol, University of Bristol, Whitson Street, Bristol BS1 3NY
| | | | | | | | | |
Collapse
|
24
|
Finch AR, Caunt CJ, Armstrong SP, McArdle CA. Plasma membrane expression of gonadotropin-releasing hormone receptors: regulation by peptide and nonpeptide antagonists. Mol Endocrinol 2009; 24:423-35. [PMID: 20009083 DOI: 10.1210/me.2009-0343] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gonadotropin-releasing hormone acts via cell surface receptors but most human (h) GnRH receptors (GnRHRs) are intracellular. A membrane-permeant nonpeptide antagonist [(2S)-2-[5-[2-(2-axabicyclo[2.2.2]oct-2-yl)-1,1-dimethy-2-oxoethyl]-2-(3,5-dimethylphenyl)-1H-indol-3-yl]-N-(2-pyridin-4-ylethyl)propan-1-amine (IN3)] increases hGnRHR expression at the surface, apparently by facilitating its exit from the endoplasmic reticulum. Here we have quantified GnRHR by automated imaging in HeLa cells transduced with adenovirus expressing hemagglutinin-tagged GnRHR. Consistent with an intracellular site of action, IN3 increases cell surface hGnRHR, and this effect is not blocked or mimicked by membrane-impermeant peptide antagonists [Ac-D2Nal-D4Cpa-D3Pal-Ser-Tyr-d-Cit-Leu-Arg-Pro-d-Ala-NH(2) (cetrorelix) and antide]. However, when the C-terminal tail of a Xenopus (X) GnRHR was added (h.XGnRHR) to increase expression, both peptides further increased cell surface GnRHR. Cetrorelix also synergized with IN3 to increase expression of hGnRHR and a G-protein coupling-deficient mutant (A261K-hGnRHR). Cetrorelix also increased cell surface expression of hGnRHR, h.XGnRHR, and mouse GnRHR in gonadotrope-lineage LbetaT2 cells, and in HeLa cells it slowed h.XGnRHR internalization (measured by receptor-mediated antihemagglutinin uptake). Thus cetrorelix has effects other than GnRHR blockade; it acts as an inverse agonist in internalization assays, supporting the potential importance of ligand-biased efficacy at GnRHR. We also developed an imaging assay for GnRH function based on Ca(2+)-dependent nuclear translocation of a nuclear factor of activated T cells reporter. Using this in HeLa and LbetaT2 cells, IN3 and cetrorelix behaved as competitive antagonists when coincubated with GnRH, and long-term pretreatment (16 h) with IN3 reduced its effectiveness as an inhibitor whereas pretreatment with cetrorelix increased its inhibitory effect. This distinction between peptide and nonpeptide antagonists may prove important for therapeutic applications of GnRH antagonists.
Collapse
Affiliation(s)
- Ann R Finch
- Laboratory for Integrative Neuroscience, Department of Clinical Sciences at South Bristol, Whitson Street, Bristol BS1 3NY, United Kingdom
| | | | | | | |
Collapse
|
25
|
Abstract
In the past decade, high-content screening has become a highly developed approach to obtaining richly descriptive quantitative phenotypic data using automated microscopy. From early use in drug screening, the technique has evolved to embrace a diverse range of applications in both academic and industrial sectors and is now widely recognized as providing an efficient and effective approach to large-scale programs investigating cell biology in situ and in context.
Collapse
Affiliation(s)
- Nick Thomas
- GE Healthcare, Whitchurch, Cardiff, United Kingdom,
| |
Collapse
|
26
|
Finch AR, Sedgley KR, Armstrong SP, Caunt CJ, McArdle CA. Trafficking and signalling of gonadotrophin-releasing hormone receptors: an automated imaging approach. Br J Pharmacol 2009; 159:751-60. [PMID: 19888967 DOI: 10.1111/j.1476-5381.2009.00413.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Gonadotrophin-releasing hormone (GnRH) is a neuropeptide that mediates central control of reproduction by stimulating gonadotrophin secretion from the pituitary. It acts via 7 transmembrane region (7TM) receptors that lack C-terminal tails, regions that for many 7TM receptors, are necessary for agonist-induced phosphorylation and arrestin binding as well as arrestin-dependent desensitization, internalization and signalling. Recent work has revealed that human GnRH receptors (GnRHR) are poorly expressed at the cell surface. This apparently reflects inefficient exit from the endoplasmic reticulum, which is thought to be increased by pharmacological chaperones (non-peptide GnRHR antagonists that increase cell surface GnRHR expression) or reduced by point mutations that further impair GnRHR trafficking and thereby cause infertility. Here, we review recent work in this field, with emphasis on the use of semi-automated imaging to interrogate compartmentalization and trafficking of these unique 7TM receptors.
Collapse
Affiliation(s)
- A R Finch
- University of Bristol, Labs. for Integrative Neuroscience and Endocrinology, Department of Clinical Sciences at South Bristol, Bristol, UK
| | | | | | | | | |
Collapse
|
27
|
Hazell GGJ, Yao ST, Roper JA, Prossnitz ER, O'Carroll AM, Lolait SJ. Localisation of GPR30, a novel G protein-coupled oestrogen receptor, suggests multiple functions in rodent brain and peripheral tissues. J Endocrinol 2009; 202:223-36. [PMID: 19420011 PMCID: PMC2710976 DOI: 10.1677/joe-09-0066] [Citation(s) in RCA: 288] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 05/01/2009] [Accepted: 05/05/2009] [Indexed: 12/11/2022]
Abstract
Recently, the G protein-coupled receptor GPR30 has been identified as a novel oestrogen receptor (ER). The distribution of the receptor has been thus far mapped only in the rat central nervous system. This study was undertaken to map the distribution of GPR30 in the mouse brain and rodent peripheral tissues. Immunohistochemistry using an antibody against GPR30 revealed high levels of GPR30 immunoreactivity (ir) in the forebrain (e.g. cortex, hypothalamus and hippocampus), specific nuclei of the midbrain (e.g. the pontine nuclei and locus coeruleus) and the trigeminal nuclei and cerebellum Purkinje layer of the hindbrain in the adult mouse brain. In the rat and mouse periphery, GPR30-ir was detected in the anterior, intermediate and neural lobe of the pituitary, adrenal medulla, renal pelvis and ovary. In situ hybridisation histochemistry using GPR30 riboprobes, revealed intense hybridisation signal for GPR30 in the paraventricular nucleus and supraoptic nucleus (SON) of the hypothalamus, anterior and intermediate lobe of the pituitary, adrenal medulla, renal pelvis and ovary of both rat and mouse. Double immunofluorescence revealed GPR30 was present in both oxytocin and vasopressin neurones of the paraventricular nucleus and SON of the rat and mouse brain. The distribution of GPR30 is distinct from the other traditional ERs and offers an additional way in which oestrogen may mediate its effects in numerous brain regions and endocrine systems in the rodent.
Collapse
|
28
|
Finch AR, Caunt CJ, Armstrong SP, McArdle CA. Agonist-induced internalization and downregulation of gonadotropin-releasing hormone receptors. Am J Physiol Cell Physiol 2009; 297:C591-600. [PMID: 19587220 PMCID: PMC2740399 DOI: 10.1152/ajpcell.00166.2009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gonadotropin-releasing hormone (GnRH) acts via seven transmembrane receptors to stimulate gonadotropin secretion. Sustained stimulation desensitizes GnRH receptor (GnRHR)-mediated gonadotropin secretion, and this underlies agonist use in hormone-dependent cancers. Since type I mammalian GnRHR do not desensitize, agonist-induced internalization and downregulation may underlie desensitization of GnRH-stimulated gonadotropin secretion; however, research focus has recently shifted to anterograde trafficking, with the finding that human (h)GnRHR are mostly intracellular. Moreover, there is little direct evidence for agonist-induced trafficking of hGnRHR, and whether or not type I mammalian GnRHR show agonist-induced internalization is controversial. Here we use automated imaging to monitor expression and internalization of hemagglutinin (HA)-tagged hGnRHRs, mouse (m) GnRHR, Xenopus (X) GnRHRs, and chimeric receptors (hGnRHR with added XGnRHR COOH tails, h.XGnRHR) expressed by adenoviral transduction in HeLa cells. We find that agonists stimulate downregulation and/or internalization of mGnRHR and XGnRHR, that GnRH stimulates trafficking of hGnRHR and can stimulate internalization or downregulation of hGnRHR when steps are taken to increase cell surface expression (addition of the XGnRHR COOH tail or pretreatment with pharmacological chaperone). Agonist effects on internalization (of h.XGnRHR) and downregulation (of hGnRHR and h.XGnRHR) were not mimicked by a peptide antagonist and were prevented by a mutation that prevents GnRHR signaling, demonstrating dependence on receptor signaling as well as agonist occupancy. Thus agonist-induced internalization and downregulation of type I mammalian GnRHR occurs in HeLa cells, and we suggest that the high throughput imaging systems described here will facilitate study of the molecular mechanisms involved.
Collapse
Affiliation(s)
- Ann R Finch
- Univ. of Bristol, Laboratory for Integrative Neuroscience, Bristol, BS1 3NY, UK
| | | | | | | |
Collapse
|
29
|
Xing Y, Nakamura Y, Rainey WE. G protein-coupled receptor expression in the adult and fetal adrenal glands. Mol Cell Endocrinol 2009; 300:43-50. [PMID: 19027826 PMCID: PMC2679220 DOI: 10.1016/j.mce.2008.10.036] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 10/22/2008] [Accepted: 10/22/2008] [Indexed: 10/21/2022]
Abstract
Hormonal regulation of adrenal function occurs primarily through G protein-coupled receptors (GPCR), which may play different roles in fetal vs. adult adrenal glands. In this study, we compared the transcript levels of GPCR between fetal and adult adrenal and found that gonadotropin-releasing hormone receptor (GnRHR), latrophilin 3 receptor, G protein-coupled receptor 37, angiotensin II receptor type 2, latrophilin 2 receptor and melanocortin receptor were expressed at significantly higher levels in fetal adrenal. High GnRHR protein expression was also detected in fetal adrenal using immunohistochemical analysis. To define potential ligand sources for fetal adrenal GnRHR, we demonstrated that GnRH1 mRNA was expressed at high levels in the placenta, while fetal adrenal had high expression of GnRH2. In summary, certain GPCR particularly GnRHR were highly expressed in fetal adrenal and the expression of GnRH mRNA in the placenta and the fetal adrenal raises the possibility of endocrine and/or paracrine/autocrine influences on fetal adrenal function. However, the exact function of GnRHR in fetal adrenal remains to be determined.
Collapse
MESH Headings
- Adrenal Glands/cytology
- Adrenal Glands/physiology
- Female
- Fetus/anatomy & histology
- Fetus/physiology
- Gene Expression Regulation, Developmental
- Humans
- Oligonucleotide Array Sequence Analysis
- Pregnancy
- Receptor, Angiotensin, Type 2/genetics
- Receptor, Angiotensin, Type 2/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, LHRH/genetics
- Receptors, LHRH/metabolism
- Receptors, Melanocortin/genetics
- Receptors, Melanocortin/metabolism
- Receptors, Peptide/genetics
- Receptors, Peptide/metabolism
Collapse
Affiliation(s)
- Yewei Xing
- Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912
| | - Yasuhiro Nakamura
- Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912
| | - William E. Rainey
- Department of Physiology, Medical College of Georgia, Augusta, Georgia 30912
| |
Collapse
|
30
|
Skinner DC, Albertson AJ, Navratil A, Smith A, Mignot M, Talbott H, Scanlan-Blake N. Effects of gonadotrophin-releasing hormone outside the hypothalamic-pituitary-reproductive axis. J Neuroendocrinol 2009; 21:282-92. [PMID: 19187469 PMCID: PMC2669307 DOI: 10.1111/j.1365-2826.2009.01842.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gonadotrophin-releasing hormone (GnRH) is a hypothalamic decapeptide with an undisputed role as a primary regulator of gonadal function. It exerts this regulation by controlling the release of gonadotrophins. However, it is becoming apparent that GnRH may have a variety of other vital roles in normal physiology. A reconsideration of the potential widespread action that this traditional reproductive hormone exerts may lead to the generation of novel therapies and provide insight into seemingly incongruent outcomes from current treatments using GnRH analogues to combat diseases such as prostate cancer.
Collapse
Affiliation(s)
- D C Skinner
- Neurobiology Program and Department of Zoology and Physiology, University of Wyoming, Laramie, WY 82071, USA.
| | | | | | | | | | | | | |
Collapse
|