1
|
Schally AV, Theodoropoulos G, Sha W, Vidaurre I, Wangpaichitr M. A 50-year journey in the development of treatment for benign prostatic hyperplasia. NPJ AGING 2025; 11:41. [PMID: 40410203 PMCID: PMC12102307 DOI: 10.1038/s41514-025-00231-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/29/2025] [Indexed: 05/25/2025]
Abstract
Recent research underscores the crucial role of hormone regulation in benign prostatic hyperplasia (BPH) and the therapeutic promise of growth hormone-releasing hormone (GH-RH) antagonists. BPH incidence in aging men doubled over three decades, driven by prostatic enlargement and lower urinary tract symptoms (LUTS). Aging-related changes in GH-RH and luteinizing hormone-releasing hormone (LH-RH) biology promote BPH through hormonal and inflammatory processes. Traditional therapies provide symptomatic relief but often fail to prevent progression. This review explores the 50-year extensive development of LH-RH and GH-RH peptide analogs from discovery to delivery and their potential in BPH treatment. In preclinical studies, GH-RH antagonists reduced prostate volume, improved LUTS, and modulated inflammation mediated by NF-κB and IGF-I. Clinical trials are needed to validate antagonist efficacy and safety. Given BPH's public health impact among the aged, and especially among aging Veterans, integrating GH-RH antagonists into management strategies may offer precision-based therapeutic advancements.
Collapse
Affiliation(s)
- Andrew V Schally
- Endocrine and Polypeptide Institute, Veterans Affairs Healthcare System, Miami, FL, USA
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service, Miami, FL, USA
- South Florida VA Foundation for Research and Education, Veterans Affairs Healthcare System, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Medicine, Divisions of Oncology and Endocrinology, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - George Theodoropoulos
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service, Miami, FL, USA
- South Florida VA Foundation for Research and Education, Veterans Affairs Healthcare System, Miami, FL, USA
| | - Wei Sha
- Endocrine and Polypeptide Institute, Veterans Affairs Healthcare System, Miami, FL, USA
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irving Vidaurre
- Endocrine and Polypeptide Institute, Veterans Affairs Healthcare System, Miami, FL, USA
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service, Miami, FL, USA
| | - Medhi Wangpaichitr
- Endocrine and Polypeptide Institute, Veterans Affairs Healthcare System, Miami, FL, USA.
- Department of Veterans Affairs, Miami VA Healthcare System, Research Service, Miami, FL, USA.
- South Florida VA Foundation for Research and Education, Veterans Affairs Healthcare System, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Surgery, Division of Thoracic Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
2
|
Desaulniers AT, White BR. Role of gonadotropin-releasing hormone 2 and its receptor in human reproductive cancers. Front Endocrinol (Lausanne) 2024; 14:1341162. [PMID: 38260130 PMCID: PMC10800933 DOI: 10.3389/fendo.2023.1341162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Gonadotropin-releasing hormone (GnRH1) and its receptor (GnRHR1) drive reproduction by regulating gonadotropins. Another form, GnRH2, and its receptor (GnRHR2), also exist in mammals. In humans, GnRH2 and GnRHR2 genes are present, but coding errors in the GnRHR2 gene are predicted to hinder full-length protein production. Nonetheless, mounting evidence supports the presence of a functional GnRHR2 in humans. GnRH2 and its receptor have been identified throughout the body, including peripheral reproductive tissues like the ovary, uterus, breast, and prostate. In addition, GnRH2 and its receptor have been detected in a wide number of reproductive cancer cells in humans. Notably, GnRH2 analogues have potent anti-proliferative, pro-apoptotic, and/or anti-metastatic effects on various reproductive cancers, including endometrial, breast, placental, ovarian, and prostate. Thus, GnRH2 is an emerging target to treat human reproductive cancers.
Collapse
Affiliation(s)
- Amy T. Desaulniers
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Brett R. White
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
3
|
Kim JW, Park M, Kim S, Lim SC, Kim HS, Kang KW. Anti-metastatic effect of GV1001 on prostate cancer cells; roles of GnRHR-mediated Gαs-cAMP pathway and AR-YAP1 axis. Cell Biosci 2021. [PMID: 34743733 DOI: 10.1186/s13578-021-00704-3.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gonadotropin-releasing hormone receptor (GnRHR) transmits its signal via two major Gα-proteins, primarily Gαq and Gαi. However, the precise mechanism underlying the functions of Gαs signal in prostate cancer cells is still unclear. We have previously identified that GV1001, a fragment of the human telomerase reverse transcriptase, functions as a biased GnRHR ligand to selectively stimulate the Gαs/cAMP pathway. Here, we tried to reveal the potential mechanisms of which GV1001-stimulated Gαs-cAMP signaling pathway reduces the migration and metastasis of prostate cancer (PCa) cells. METHODS The expression of epithelial-mesenchymal transition (EMT)-related genes was measured by western-blotting and spheroid formation on ultra-low attachment plate was detected after GV1001 treatment. In vivo Spleen-liver metastasis mouse model was used to explore the inhibitory effect of GV1001 on metastatic ability of PCa and the transwell migration assay was performed to identify whether GV1001 had a suppressive effect on cell migration in vitro. In order to demonstrate the interaction between androgen receptor (AR) and YAP1, co-immunoprecipitation (co-IP), immunofluorescence (IF) staining, chromatin immunoprecipitation (ChIP) were performed in LNCaP cells with and without GV1001 treatment. RESULTS GV1001 inhibited expression of EMT-related genes and spheroid formation. GV1001 also suppressed in vivo spleen-liver metastasis of LNCaP cells as well as cell migration in vitro. GV1001 enhanced the phosphorylation of AR and transcription activity of androgen response element reporter gene through cAMP/protein kinase A pathway. Moreover, GV1001 increased Ser-127 phosphorylation of YAP1 and its ubiquitination, and subsequently decreased the levels of AR-YAP1 binding in the promoter region of the CTGF gene. In contrast, both protein and mRNA levels of NKX3.1 known for tumor suppressor gene and AR-coregulator were upregulated by GV1001 in LNCaP cells. YAP1 knockout using CRISPR/Cas9 significantly suppressed the migration ability of LNCaP cells, and GV1001 did not affect the cell migration of YAP1-deficient LNCaP cells. On the contrary, cell migration was more potentiated in LNCaP cells overexpressing YAP5SA, a constitutively active form of YAP1, which was not changed by GV1001 treatment. CONCLUSIONS Overall, this study reveals an essential role of AR-YAP1 in the regulation of PCa cell migration, and provides evidence that GV1001 could be a novel GnRHR ligand to inhibit metastasis of PCa via the Gαs/cAMP pathway.
Collapse
Affiliation(s)
- Ji Won Kim
- Division of Hematology and Medical Oncology, University of California, San Francisco, CA, 94143, USA
| | - Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suntae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Chul Lim
- Department of Pathology, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hyung Shik Kim
- College of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
4
|
Kim JW, Park M, Kim S, Lim SC, Kim HS, Kang KW. Anti-metastatic effect of GV1001 on prostate cancer cells; roles of GnRHR-mediated Gαs-cAMP pathway and AR-YAP1 axis. Cell Biosci 2021; 11:191. [PMID: 34743733 PMCID: PMC8574053 DOI: 10.1186/s13578-021-00704-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background Gonadotropin-releasing hormone receptor (GnRHR) transmits its signal via two major Gα-proteins, primarily Gαq and Gαi. However, the precise mechanism underlying the functions of Gαs signal in prostate cancer cells is still unclear. We have previously identified that GV1001, a fragment of the human telomerase reverse transcriptase, functions as a biased GnRHR ligand to selectively stimulate the Gαs/cAMP pathway. Here, we tried to reveal the potential mechanisms of which GV1001-stimulated Gαs-cAMP signaling pathway reduces the migration and metastasis of prostate cancer (PCa) cells. Methods The expression of epithelial-mesenchymal transition (EMT)-related genes was measured by western-blotting and spheroid formation on ultra-low attachment plate was detected after GV1001 treatment. In vivo Spleen-liver metastasis mouse model was used to explore the inhibitory effect of GV1001 on metastatic ability of PCa and the transwell migration assay was performed to identify whether GV1001 had a suppressive effect on cell migration in vitro. In order to demonstrate the interaction between androgen receptor (AR) and YAP1, co-immunoprecipitation (co-IP), immunofluorescence (IF) staining, chromatin immunoprecipitation (ChIP) were performed in LNCaP cells with and without GV1001 treatment. Results GV1001 inhibited expression of EMT-related genes and spheroid formation. GV1001 also suppressed in vivo spleen-liver metastasis of LNCaP cells as well as cell migration in vitro. GV1001 enhanced the phosphorylation of AR and transcription activity of androgen response element reporter gene through cAMP/protein kinase A pathway. Moreover, GV1001 increased Ser-127 phosphorylation of YAP1 and its ubiquitination, and subsequently decreased the levels of AR-YAP1 binding in the promoter region of the CTGF gene. In contrast, both protein and mRNA levels of NKX3.1 known for tumor suppressor gene and AR-coregulator were upregulated by GV1001 in LNCaP cells. YAP1 knockout using CRISPR/Cas9 significantly suppressed the migration ability of LNCaP cells, and GV1001 did not affect the cell migration of YAP1-deficient LNCaP cells. On the contrary, cell migration was more potentiated in LNCaP cells overexpressing YAP5SA, a constitutively active form of YAP1, which was not changed by GV1001 treatment. Conclusions Overall, this study reveals an essential role of AR-YAP1 in the regulation of PCa cell migration, and provides evidence that GV1001 could be a novel GnRHR ligand to inhibit metastasis of PCa via the Gαs/cAMP pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00704-3.
Collapse
Affiliation(s)
- Ji Won Kim
- Division of Hematology and Medical Oncology, University of California, San Francisco, CA, 94143, USA
| | - Miso Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Suntae Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Sung Chul Lim
- Department of Pathology, College of Medicine, Chosun University, Gwangju, 61452, Republic of Korea
| | - Hyung Shik Kim
- College of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Keon Wook Kang
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
5
|
Fontana F, Limonta P. Dissecting the Hormonal Signaling Landscape in Castration-Resistant Prostate Cancer. Cells 2021; 10:1133. [PMID: 34067217 PMCID: PMC8151003 DOI: 10.3390/cells10051133] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Understanding the molecular mechanisms underlying prostate cancer (PCa) progression towards its most aggressive, castration-resistant (CRPC) stage is urgently needed to improve the therapeutic options for this almost incurable pathology. Interestingly, CRPC is known to be characterized by a peculiar hormonal landscape. It is now well established that the androgen/androgen receptor (AR) axis is still active in CRPC cells. The persistent activity of this axis in PCa progression has been shown to be related to different mechanisms, such as intratumoral androgen synthesis, AR amplification and mutations, AR mRNA alternative splicing, increased expression/activity of AR-related transcription factors and coregulators. The hypothalamic gonadotropin-releasing hormone (GnRH), by binding to its specific receptors (GnRH-Rs) at the pituitary level, plays a pivotal role in the regulation of the reproductive functions. GnRH and GnRH-R are also expressed in different types of tumors, including PCa. Specifically, it has been demonstrated that, in CRPC cells, the activation of GnRH-Rs is associated with a significant antiproliferative/proapoptotic, antimetastatic and antiangiogenic activity. This antitumor activity is mainly mediated by the GnRH-R-associated Gαi/cAMP signaling pathway. In this review, we dissect the molecular mechanisms underlying the role of the androgen/AR and GnRH/GnRH-R axes in CRPC progression and the possible therapeutic implications.
Collapse
Affiliation(s)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milano, Italy;
| |
Collapse
|
6
|
Role of Gonadotropin-Releasing Hormone (GnRH) in Ovarian Cancer. Cells 2021; 10:cells10020437. [PMID: 33670761 PMCID: PMC7922220 DOI: 10.3390/cells10020437] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/28/2021] [Accepted: 02/17/2021] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus–pituitary–gonadal (HPG) axis is the endocrine regulation system that controls the woman’s cycle. The gonadotropin-releasing hormone (GnRH) plays the central role. In addition to the gonadotrophic cells of the pituitary, GnRH receptors are expressed in other reproductive organs, such as the ovary and in tumors originating from the ovary. In ovarian cancer, GnRH is involved in the regulation of proliferation and metastasis. The effects on ovarian tumors can be indirect or direct. GnRH acts indirectly via the HPG axis and directly via GnRH receptors on the surface of ovarian cancer cells. In this systematic review, we will give an overview of the role of GnRH in ovarian cancer development, progression and therapy.
Collapse
|
7
|
Mugami S, Dobkin-Bekman M, Rahamim-Ben Navi L, Naor Z. Differential roles of PKC isoforms (PKCs) in GnRH stimulation of MAPK phosphorylation in gonadotrope derived cells. Mol Cell Endocrinol 2018; 463:97-105. [PMID: 28392410 DOI: 10.1016/j.mce.2017.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/30/2022]
Abstract
The role of protein kinase C (PKC) isoforms (PKCs) in GnRH-stimulated MAPK [ERK1/2, JNK1/2 and p38) phosphorylation was examined in gonadotrope derived cells. GnRH induced a protracted activation of ERK1/2 and a slower and more transient activation of JNK1/2 and p38MAPK. Gonadotropes express conventional PKCα and PKCβII, novel PKCδ, PKCε and PKCθ, and atypical PKC-ι/λ. The use of green fluorescent protein (GFP)-PKCs constructs revealed that GnRH induced rapid translocation of PKCα and PKCβII to the plasma membrane, followed by their redistribution to the cytosol. PKCδ and PKCε localized to the cytoplasm and Golgi, followed by the rapid redistribution by GnRH of PKCδ to the perinuclear zone and of PKCε to the plasma membrane. The use of dominant negatives for PKCs and peptide inhibitors for the receptors for activated C kinase (RACKs) has revealed differential role for PKCα, PKCβII, PKCδ and PKCε in ERK1/2, JNK1/2 and p38MAPK phosphorylation in a ligand-and cell context-dependent manner. The paradoxical findings that PKCs activated by GnRH and PMA play a differential role in MAPKs phosphorylation may be explained by persistent vs. transient redistribution of selected PKCs or redistribution of a given PKC to the perinuclear zone vs. the plasma membrane. Thus, we have identified the PKCs involved in GnRH stimulated MAPKs phosphorylation in gonadotrope derived cells. Once activated, the MAPKs will mediate the transcription of the gonadotropin subunits and GnRH receptor genes.
Collapse
Affiliation(s)
- Shany Mugami
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Masha Dobkin-Bekman
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Liat Rahamim-Ben Navi
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Zvi Naor
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel.
| |
Collapse
|
8
|
Targeting luteinizing hormone-releasing hormone: A potential therapeutics to treat gynecological and other cancers. J Control Release 2018; 269:277-301. [DOI: 10.1016/j.jconrel.2016.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 01/05/2023]
|
9
|
Corona SP, Roviello G, Strina C, Milani M, Allevi G, Aguggini S, Zanoni D, Generali D. Could gonadotropin-releasing hormone analogs be helpful in the treatment of triple-negative breast cancer? Future Oncol 2017; 13:2473-2477. [PMID: 29121807 DOI: 10.2217/fon-2017-0272] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/27/2017] [Indexed: 11/21/2022] Open
Abstract
AIM Treatment of triple-negative breast cancer (TNBC) imposes great challenges, due to a lack of molecular targets. While use of gonadotropin-releasing hormone (GnRH) analogs has been validated in ER-positive breast cancer, this option has not been investigated in TNBC, even though a significant portion of these tumors upregulate GnRH receptors. We performed a meta-analysis of the literature to evaluate the effect of GnRH analogs in TNBC. METHODS Four studies were included in this study. RESULTS We detected a non-significant improvement in overall survival with GnRH analogs, while progression-free survival was unchanged. DISCUSSION The majority of the trials evaluated in this analysis were designed to test efficacy of GnRH analogs in preventing premature ovarian failure. This may represent a limitation of our study as these trials were not specifically designed to detect differences in survival outcome measures. CONCLUSION Our results suggest that GnRH analogs may be useful as a targeted therapy in TNBC. Randomized prospective clinical trials are needed to investigate this hypothesis in the clinic.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Peter Mac Callum Cancer Centre, Moorabbin Campus, 823-865 Centre Rd, Bentleigh East, VIC 3165, Australia
| | - Giandomenico Roviello
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, Trieste, 34129, Italy
- Department of Oncology, Medical Oncology Unit, San Donato Hospital, Arezzo, 52100, Italy
| | - Carla Strina
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| | - Manuela Milani
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| | - Giovanni Allevi
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| | - Sergio Aguggini
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| | - Daniele Zanoni
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| | - Daniele Generali
- Department of Medical, Surgery and Health Sciences, University of Trieste, Piazza Ospitale 1, Trieste, 34129, Italy
- UO Multidisciplinare di Patologia Mammaria, US Terapia Molecolare e Farmacogenomica, ASST Cremona, Viale Concordia 1, Cremona, 26100, Italy
| |
Collapse
|
10
|
Schally AV, Block NL, Rick FG. Discovery of LHRH and development of LHRH analogs for prostate cancer treatment. Prostate 2017; 77:1036-1054. [PMID: 28449236 DOI: 10.1002/pros.23360] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 04/06/2017] [Indexed: 01/06/2023]
Abstract
The discovery, isolation, elucidation of structure, synthesis, and initial testing of the neuropeptide hypothalamic luteinizing hormone-releasing hormone (LHRH), which regulates reproduction, is briefly described. The design, synthesis, and experimental and clinical testing of agonistic analogs of LHRH is extensively reviewed focusing on the development of new methods for the treatment of prostate cancer. Subsequent development of antagonistic analogs of LHRH is then faithfully recounted with special emphasis on therapy of prostate cancer and BPH. The concepts of targeted therapy to peptide receptors on tumors are re-examined and the development of the cytotoxic analogs of LHRH and their status is reviewed. The endeavor to develop better therapies for prostate cancer, based on LHRH analogs, guided much of our work.
Collapse
Affiliation(s)
- Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Division of Endocrinology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Norman L Block
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
- Division of Hematology/Oncology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, Florida
- Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida
| |
Collapse
|
11
|
Crawford ED, Schally AV, Pinthus JH, Block NL, Rick FG, Garnick MB, Eckel RH, Keane TE, Shore ND, Dahdal DN, Beveridge TJR, Marshall DC. The potential role of follicle-stimulating hormone in the cardiovascular, metabolic, skeletal, and cognitive effects associated with androgen deprivation therapy. Urol Oncol 2017; 35:183-191. [PMID: 28325650 DOI: 10.1016/j.urolonc.2017.01.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/20/2017] [Accepted: 01/24/2017] [Indexed: 11/25/2022]
Abstract
PURPOSE To explore how follicle-stimulating hormone (FSH) may contribute to cardiovascular, metabolic, skeletal, and cognitive events in men treated for prostate cancer, with various forms of androgen deprivation therapy (ADT). MATERIALS AND METHODS A colloquium of prostate cancer experts was convened in May 2015, to discuss the role of FSH in the development of unwanted effects associated with ADT. Subsequently, a literature review (Medline, PubMed, and relevant congress abstract databases) was performed to further explore and evaluate the collected evidence. RESULTS It has become evident that, in the setting of ADT, FSH can promote the development of atherosclerotic plaque formation, metabolic syndrome, and insulin resistance. Data also suggest that FSH is an important mediator of bone remodeling, particularly bone resorption, and thereby increases the risk for bone fracture. Additional evidence implicates a role for FSH in bone metastasis as well. The influence of FSH on ADT-induced cognitive deficits awaits further elucidation; however, the possibility that FSH may be involved therein cannot be ruled out. CONCLUSIONS The widespread molecular and physiological consequences of FSH system activation in normal and pathological conditions are becoming better understood. Progress in this area has been achieved by the development of additional investigative and clinical measures to better evaluate specific adverse effects. More research is needed on FSH function in the development of cancer as well as its association with cardiovascular, metabolic, musculoskeletal, and cognitive effects in ADT.
Collapse
Affiliation(s)
- E David Crawford
- Department of Urologic Oncology, School of Medicine, University of Colorado, Denver, Denver, CO.
| | - Andrew V Schally
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Pathology, University of Miami School of Medicine, Miami, FL; Department of Medicine, University of Miami School of Medicine, Miami, FL
| | - Jehonathan H Pinthus
- Department of Surgery, Juravinski Cancer Centre, McMaster University, Hamilton, Ontario, Canada
| | - Norman L Block
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Pathology, University of Miami School of Medicine, Miami, FL; Department of Medicine, University of Miami School of Medicine, Miami, FL
| | - Ferenc G Rick
- Endocrine, Polypeptide and Cancer Institute, Miami Veterans Affairs Medical Center, Miami, FL; Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| | - Marc B Garnick
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Robert H Eckel
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado, Denver, CO
| | - Thomas E Keane
- Department of Urology, Medical University of South Carolina, Charleston, SC
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC
| | | | | | | |
Collapse
|
12
|
Mugami S, Kravchook S, Rahamim-Ben Navi L, Seger R, Naor Z. Differential roles of PKC isoforms (PKCs) and Ca 2+ in GnRH and phorbol 12-myristate 13-acetate (PMA) stimulation of p38MAPK phosphorylation in immortalized gonadotrope cells. Mol Cell Endocrinol 2017; 439:141-154. [PMID: 27810601 DOI: 10.1016/j.mce.2016.10.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/25/2016] [Accepted: 10/28/2016] [Indexed: 10/20/2022]
Abstract
We examined the role of PKCs and Ca2+ in GnRH-stimulated p38MAPK phosphorylation in the gonadotrope derived αT3-1 and LβT2 cell lines. GnRH induced a slow and rapid increase in p38MAPK phosphorylation in αT3-1 and LβT2 cells respectively, while PMA gave a slow response. The use of dominant negatives for PKCs and peptide inhibitors for the receptors for activated C kinase (RACKs), has revealed differential role for PKCα, PKCβII, PKCδ and PKCε in p38MAPK phosphorylation in a ligand-and cell context-dependent manner. The paradoxical findings that PKCs activated by GnRH and PMA play a differential role in p38MAPK phosphorylation may be explained by differential localization of the PKCs. Basal, GnRH- and PMA- stimulation of p38MAPK phosphorylation in αT3-1 cells is mediated by Ca2+ influx via voltage-gated Ca2+ channels and Ca2+ mobilization, while in the differentiated LβT2 gonadotrope cells it is mediated only by Ca2+ mobilization. p38MAPK resides in the cell membrane and is relocated to the nucleus by GnRH (∼5 min). Thus, we have identified the PKCs and the Ca2+ pools involved in GnRH stimulated p38MAPK phosphorylation.
Collapse
Affiliation(s)
- Shany Mugami
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Shani Kravchook
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Liat Rahamim-Ben Navi
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | - Rony Seger
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Zvi Naor
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel.
| |
Collapse
|
13
|
Gründker C, Emons G. The Role of Gonadotropin-Releasing Hormone in Cancer Cell Proliferation and Metastasis. Front Endocrinol (Lausanne) 2017; 8:187. [PMID: 28824547 PMCID: PMC5543040 DOI: 10.3389/fendo.2017.00187] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022] Open
Abstract
In several human malignant tumors of the urogenital tract, including cancers of the endometrium, ovary, urinary bladder, and prostate, it has been possible to identify expression of gonadotropin-releasing hormone (GnRH) and its receptor as part of an autocrine system, which regulates cell proliferation. The expression of GnRH receptor has also been identified in breast cancers and non-reproductive cancers such as pancreatic cancers and glioblastoma. Various investigators have observed dose- and time-dependent growth inhibitory effects of GnRH agonists in cell lines derived from these cancers. GnRH antagonists have also shown marked growth inhibitory effects on most cancer cell lines. This indicates that in the GnRH system in cancer cells, there may not be a dichotomy between GnRH agonists and antagonists. The well-known signaling mechanisms of the GnRH receptor, which are present in pituitary gonadotrophs, are not involved in forwarding the antiproliferative effects of GnRH analogs in cancer cells. Instead, the GnRH receptor activates a phosphotyrosine phosphatase (PTP) and counteracts with the mitogenic signal transduction of growth factor receptors, which results in a reduction of cancer cell proliferation. The PTP activation, which is induced by GnRH, also inhibits G-protein-coupled estrogen receptor 1 (GPER), which is a membrane-bound receptor for estrogens. GPER plays an important role in breast cancers, which do not express the estrogen receptor α (ERα). In metastatic breast, ovarian, and endometrial cancer cells, GnRH reduces cell invasion in vitro, metastasis in vivo, and the increased expression of S100A4 and CYR61. All of these factors play important roles in epithelial-mesenchymal transition. This review will summarize the present state of knowledge about the GnRH receptor and its signaling in human cancers.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | - Günter Emons
- Department of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| |
Collapse
|
14
|
Terashima R, Laoharatchatathanin T, Kurusu S, Kawaminami M. Augmentation of gonadotropin-releasing hormone receptor expression in the post-lactational mammary tissues of rats. J Reprod Dev 2016; 62:495-499. [PMID: 27349532 PMCID: PMC5081737 DOI: 10.1262/jrd.2016-035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is a neurohormone of the hypothalamus controlling pituitary gonadotropin secretion and hence gametogenesis. While it has also been believed that GnRH is synthesized and functions in various peripheral tissues, the expression of GnRH receptor (GnRH-R) in peripheral tissues is not well-described. We previously found that annexin A5, which is increased in the pituitary gonadotropes by GnRH, is dramatically increased in rat mammary epithelial cells after weaning, suggesting that local GnRH is responsible for this increase. Annexin A5 is a member of the annexin family of proteins and is thought to be involved in various regulatory mechanisms, including apoptosis. In the present study, we examined GnRH-R expression in the mammary tissues after weaning. Although GnRH-R mRNA was not detected in the mammary tissues during lactation, it was dramatically increased after weaning. Forced weaning at mid-lactation (day 10) also promoted the expression of GnRH-R transcripts in mammary tissues within 2 days. Furthermore, western blotting analysis with anti-GnRH-R showed that the expression of an immuno-positive 60-kDa protein, whose size was equivalent to that of rat GnRH-R, was confirmed to increase after weaning. These findings clarified the induction of GnRH-R in the mammary tissues after weaning and suggest that GnRH is involved in the involution and tissue remodeling of post-lactating rat mammary tissues.
Collapse
Affiliation(s)
- Ryota Terashima
- Laboratory of Veterinary Physiology, School of Veterinary Medicine, Kitasato University, Aomori 034-8628, Japan
| | | | | | | |
Collapse
|
15
|
Weng H, Liu F, Hu S, Li L, Wang Y. GnRH agonists induce endometrial epithelial cell apoptosis via GRP78 down-regulation. J Transl Med 2014; 12:306. [PMID: 25367189 PMCID: PMC4240864 DOI: 10.1186/s12967-014-0306-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 10/21/2014] [Indexed: 12/17/2022] Open
Abstract
Background Endometriosis is a benign chronic gynecological disease that affects women of reproductive age, characterized by the presence of functional endometrial tissues outside the uterine cavity. GnRH agonists exhibit anti-proliferative and apoptosis-enhancing activities and have long been used for the treatment of endometriosis. There is a critical need to identify the signaling modules involving GnRH agonist therapy for the treatment of endometriosis. In this study, we compared the proteomic profiles of endometriosis in patients before and after GnRH agonist therapy to identify proteins that might provide further information concerning the mechanisms underlying the functions of GnRH agonists. Methods A total of 55 protein spots with different abundances were observed using Difference Gel Electrophoresis (DIGE), and 26 of these proteins were assigned clear identities through Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Tandem Mass Spectroscopy (MALDI-TOF/TOF MS). Results We validated four of these proteins through Western blotting and immunohistochemistry using human endometrial tissue. We also characterized the effect of Leuprolide acetate (LA) on the apoptosis of eutopic endometrial epithelial cells. LA treatment significantly promoted the apoptosis of eutopic endometrial epithelial cells and inhibited the expression of the anti-apoptotic factor GRP78. GRP78 knockdown enhanced LA-induced cell apoptosis, whereas, the overexpression of GRP78 in eutopic endometrial epithelial cells suppresses LA-induced apoptosis. Conclusion These results suggest that GnRH agonists induce endometrial epithelial cell apoptosis via GRP78 down-regulation. This study might provide an important molecular framework for further evaluation of GnRH agonist therapy.
Collapse
Affiliation(s)
- Huinan Weng
- Department of Pathophysiology, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China. .,GuangDong Women and Children Hospital, Guangzhou, China.
| | - Fenghua Liu
- GuangDong Women and Children Hospital, Guangzhou, China.
| | - Shuiwang Hu
- Department of Pathophysiology, Key Laboratory of Proteomics of Guangdong Province, Southern Medical University, Guangzhou, China.
| | - Li Li
- GuangDong Women and Children Hospital, Guangzhou, China.
| | - Yifeng Wang
- ZhuJiang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
Sviridonov L, Dobkin-Bekman M, Shterntal B, Przedecki F, Formishell L, Kravchook S, Rahamim-Ben Navi L, Bar-Lev TH, Kazanietz MG, Yao Z, Seger R, Naor Z. Differential signaling of the GnRH receptor in pituitary gonadotrope cell lines and prostate cancer cell lines. Mol Cell Endocrinol 2013; 369:107-18. [PMID: 23380421 PMCID: PMC4100609 DOI: 10.1016/j.mce.2013.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
Abstract
The GnRH receptor (GnRHR) mediates the pituitary functions of GnRH, as well as its anti-proliferative effects in sex hormone-dependent cancer cells. Here we compare the signaling of GnRHR in pituitary gonadotrope cell lines vs. prostate cancer cell lines. We first noticed that the expression level of PKCα, PKCβII and PKCε is much higher in αT3-1 and LβT2 gonadotrope cell lines vs. LNCaP and DU-145 cell lines, while the opposite is seen for PKCδ. Activation of PKCα, PKCβII and PKCε by GnRH is relatively transient in αT3-1 and LβT2 gonadotrope cell lines and more prolonged in LNCaP and DU-145 cell lines. On the otherhand, the activation and re-distribution of the above PKCs by PMA was similar for both gonadotrope cell lines and prostate cancer cell lines. Activation of ERK1/2 by GnRH and PMA was robust in the gonadotrope cell lines, with a smaller effect observed in the prostate cancer cell lines. The Ca(2+) ionophore A23187 stimulated ERK1/2 in gonadotrope cell lines but not in prostate cancer cell lines. GnRH, PMA and A23187 stimulated JNK activity in gonadotrope cell lines, with a more sustained effect in prostate cancer cell lines. Sustained activation of p38 was observed for PMA and A23187 in Du-145 cells, while p38 activation by GnRH, PMA and A23187 in LβT2 cells was transient. Thus, differential expression and re-distribution of PKCs by GnRH and the transient vs. the more sustained nature of the activation of the PKC-MAPK cascade by GnRH in gonadotrope cell lines vs. prostate cancer cell lines respectively, may provide the mechanistic basis for the cell context-dependent differential biological responses observed in GnRH interaction with pituitary gonadotropes vs. prostate cancer cells.
Collapse
Affiliation(s)
- Ludmila Sviridonov
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv 69978, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
G protein-coupled receptors (GPCRs) are the largest class of integral membrane protein receptors in the human genome. We examined here the reports whether the GnRH receptor (GnRHR) interacts with a single or multiple types of G proteins. It seems that the GnRHR, as other GPCRs, alternates between various conformations and is stabilized by its ligands, other modulators and intracellular partners in selective conformations culminating in coupling with a single type or multiple G proteins in a cell- and context-specific manner.
Collapse
Affiliation(s)
- Zvi Naor
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| | | |
Collapse
|
18
|
Block M, Gründker C, Fister S, Kubin J, Wilkens L, Mueller MD, Hemmerlein B, Emons G, Günthert AR. Inhibition of the AKT/mTOR and erbB pathways by gefitinib, perifosine and analogs of gonadotropin-releasing hormone I and II to overcome tamoxifen resistance in breast cancer cells. Int J Oncol 2012; 41:1845-54. [PMID: 22922893 DOI: 10.3892/ijo.2012.1591] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 07/03/2012] [Indexed: 11/05/2022] Open
Abstract
Endocrine resistance in breast cancer remains a major clinical problem and is caused by crosstalk mechanisms of growth factor receptor cascades, such as the erbB and PI3K/AKT pathways. The possibilities a single breast cancer cell has to achieve resistance are manifold. We developed a model of 4-hydroxy-tamoxifen (OHT)‑resistant human breast cancer cell lines and compared their different expression patterns, activation of growth factor receptor pathways and compared cells by genomic hybridization (CGH). We also tested a panel of selective inhibitors of the erbB and AKT/mTOR pathways to overcome OHT resistance. OHT‑resistant MCF-7-TR and T47D-TR cells showed increased expression of HER2 and activation of AKT. T47D-TR cells showed EGFR expression and activated MAPK (ERK-1/2), whereas in resistant MCF-7-TR cells activated AKT was due to loss of CTMP expression. CGH analyses revealed remarkable aberrations in resistant sublines, which were predominantly depletions. Gefitinib inhibited erbB signalling and restored OHT sensitivity in T47D-TR cells. The AKT inhibitor perifosine restored OHT sensitivity in MCF-7-TR cells. All cell lines showed expression of receptors for gonadotropin-releasing hormone (GnRH) I and II, and analogs of GnRH-I/II restored OHT sensitivity in both resistant cell lines by inhibition of erbB and AKT signalling. In conclusion, mechanisms to escape endocrine treatment in breast cancer share similarities in expression profiling but are based on substantially different genetic aberrations. Evaluation of activated mediators of growth factor receptor cascades is helpful to predict response to specific inhibitors. Expression of GnRH-I/II receptors provides multi-targeting treatment strategies.
Collapse
Affiliation(s)
- Martin Block
- Departement of Gynecology and Obstetrics, Georg-August-University, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Li L, Fu YC, Xu JJ, Chen XC, Lin XH, Luo LL. Caloric restriction promotes the reproductive capacity of female rats via modulating the level of insulin-like growth factor-1 (IGF-1). Gen Comp Endocrinol 2011; 174:232-7. [PMID: 21945120 DOI: 10.1016/j.ygcen.2011.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2011] [Revised: 08/15/2011] [Accepted: 09/06/2011] [Indexed: 11/18/2022]
Abstract
The insulin-like growth factor-1 (IGF-1) plays an important role in the regulation of reproductive function. In the present study, we examined the effects of caloric restriction (CR) on the reproductive lifespan in rats and investigated the potential role of IGF-1. After 10 weeks of treatment, we determined the distribution of the ovarian follicles at various stages and measured the plasma level of IGF-1, luteinizing hormone (LH), follicle-stimulating hormone (FSH), and estrogen (ESG). Our results show that IGF-1 level was decreased after CR and correlated with the decrease in the levels of LH, FSH and ESG. Moreover, a higher percentage of primordial follicles and surviving follicles was observed in CR rats than in control rats (P<0.05). Immunohistochemical analysis showed that IGF-1 was extensively expressed in the cytoplasm of granulosa cells in the surviving follicles at different stages but not in the atretic follicles. Taken together, these results suggest that caloric restriction promotes the reproductive capacity of female rats via modulating the level of IGF-1, which then regulate pituitary gonadotrope cells to reduce the release of LH, FSH and ESG, and modulate follicular development.
Collapse
Affiliation(s)
- Li Li
- Department of Gynaecology and Obstetrics of the First Affiliated Hospital, Medical College, Shantou University, Shantou 515041, China.
| | | | | | | | | | | |
Collapse
|
20
|
Biochemistry, molecular biology and cell biology of gonadotropin-releasing hormone antagonists. Curr Opin Obstet Gynecol 2011; 23:238-44. [DOI: 10.1097/gco.0b013e328348a3ce] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
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
|
Dobkin-Bekman M, Rahamin-Ben Navi L, Shterntal B, Sviridonov L, Przedecki F, Naidich-Exler M, Brodie C, Seger R, Naor Z. Differential role of PKC isoforms in GnRH and phorbol 12-myristate 13-acetate activation of extracellular signal-regulated kinase and Jun N-terminal kinase. Endocrinology 2010; 151:4894-907. [PMID: 20810567 DOI: 10.1210/en.2010-0114] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH is the first key hormone of reproduction. The role of protein kinase C (PKC) isoforms in GnRH-stimulated MAPK [ERK and Jun N-terminal kinase (JNK)] was examined in the αT3-1 and LβT2 gonadotrope cells. Incubation of the cells with GnRH resulted in a protracted activation of ERK1/2 and a slower and more transient activation of JNK1/2. Gonadotropes express conventional PKCα and conventional PKCβII, novel PKCδ, novel PKCε, and novel PKCθ, and atypical PKC-ι/λ. The use of green fluorescent protein-PKC constructs revealed that GnRH induced rapid translocation of PKCα and PKCβII to the plasma membrane, followed by their redistribution to the cytosol. PKCδ and PKCε localized to the cytoplasm and Golgi, followed by the rapid redistribution by GnRH of PKCδ to the perinuclear zone and of PKCε to the plasma membrane. Interestingly, PKCα, PKCβII, and PKCε translocation to the plasma membrane was more pronounced and more prolonged in phorbol-12-myristate-13-acetate (PMA) than in GnRH-treated cells. The use of selective inhibitors and dominant-negative plasmids for the various PKCs has revealed that PKCβII, PKCδ, and PKCε mediate ERK2 activation by GnRH, whereas PKCα, PKCβII, PKCδ, and PKCε mediate ERK2 activation by PMA. Also, PKCα, PKCβII, PKCδ, and PKCε are involved in GnRH and PMA stimulation of JNK1 in a cell-context-dependent manner. We present preliminary evidence that persistent vs. transient redistribution of selected PKCs or redistribution of a given PKC to the perinuclear zone vs. the plasma membrane may dictate its selective role in ERK or JNK activation. Thus, we have described the contribution of selective PKCs to ERK and JNK activation by GnRH.
Collapse
Affiliation(s)
- Masha Dobkin-Bekman
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv 69978, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Oztürk HB, Vural B, Calışkan E, Solakoğlu S. Effect of GnRH analogues and octreotide treatment on apoptosis and the cell proliferation of endometrium adenocarcinoma cell lines. J Turk Ger Gynecol Assoc 2010; 11:131-6. [PMID: 24591918 DOI: 10.5152/jtgga.2010.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 08/12/2010] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE The aim of this study was to compare apoptotic and antiproliferative effects of gonadotropin-releasing hormone analogues and their combination with octeotide on endometrioid endometrial cancer cell lines. MATERIAL AND METHOD Women diagnosed with endometrioid adenocarcinoma at the department of Gynecology and Obstetric of Kocaeli University Medical School were included in this research. Endometrium cancer cell lines obtained from three patients were used for this study. After trypsinization in 0.5% in calcium magnesium, free phosphate buffer solution (CMFPBS) cells were seeded on glass slides in 24-well plates containing DMEM-F12 medium and 10% fetal calf serum as culture medium. Cells were incubated for 24 hours at 37ºC in 5% CO2. GnRH agonist leuprolide (Lucrin 1 μmol/L), GnRH antagonist ganirelix (Orgalutran 1 μmol/L), leuprolide with octreotide (Sandostatin 10-6 mol/L), ganirelix with octreotide and no drug were added to the wells. Apoptosis and cells proliferations were evaluated after 12, 24, 48 and 72(th) hours of incubation. The percentage of apoptotic cells was evaluated by TdT mediated biotin-dUTP nick-end labeling (TUNEL) method; cell proliferation was assessed by bromodeoxyuridine (BrdU) incorporation. RESULTS Apoptotic index in grade I EEC cell line among ganirelixoctreotide treated cells and leuprolide-octreotide combination therapy were respectively higher than the untreated control (p<0.001, p=0.001). The number of apoptotic cells in grade II EEC cell line among leuprolide-octreotide and leuprolide were significantly (p<0.001, p<0.001) higher than in controls. In grade III EEC cell line, the number of TUNEL positive cells among leuprolide, ganirelix and ganirelixoctreotide therapiy groups were significantly higher than in untreated control. Time dependent antiproliferative effect was obtained with leuprolide and leuprolide-octreotide in grade I EEC (p<0.001, p<0.001). Grade II EEC cell line is not influenced by hormonotherapies. However, the antiproliferative effect was obtained with ganirelix, leuprolide and leuprolide-octreotide in grade III cell line. CONCLUSION GnRH analogues appears to have a direct effect, enhancing the apoptotic index and decreasing the cell proliferation in endometrial adenocancer cell lines.
Collapse
Affiliation(s)
| | - Birol Vural
- Department of Obstetric and Gynecology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Eray Calışkan
- Department of Obstetric and Gynecology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Seyhun Solakoğlu
- Department of Histology and Embryology, School of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
24
|
Festuccia C, Dondi D, Piccolella M, Locatelli A, Gravina GL, Tombolini V, Motta M. Ozarelix, a fourth generation GnRH antagonist, induces apoptosis in hormone refractory androgen receptor negative prostate cancer cells modulating expression and activity of death receptors. Prostate 2010; 70:1340-9. [PMID: 20623634 DOI: 10.1002/pros.21169] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Antagonistic or agonistic analogues of gonadotropin-releasing hormone are extensively used for the treatment of advanced hormone-dependent prostate cancer. However, the majority of recurrent prostate tumors is androgen independent. This study explored the in vitro effects on DU145 and PC3 cell lines, two models of androgen-independent prostate cancer, of a fourth generation GnRH antagonist (Ozarelix). METHODS Ozarelix was added to cultures and toxicity, cell cycle modifications, cell viability and caspase activity were investigated. RESULTS Ozarelix showed antiproliferative effects and produced an accumulation of cells in G2/M cell cycle phase. Apoptosis was related with caspase-8-dependent caspase 3 activation with down-regulation of c-FLIP (L) and a sensitization to TRAIL-induced apoptosis linked also to increased expression and activity of death receptors DR4/5 and Fas. CONCLUSIONS TRAIL-resistant cancer cells can be sensitized to TRAIL by Ozarelix. This effect may be achieved by the activation of apoptotic pathway improving the therapeutic effects in androgen independent tumor cell lines. However, a better understanding of molecular mechanisms by which GnRH antagonists may act in androgen independent models is necessary.
Collapse
Affiliation(s)
- Claudio Festuccia
- Radiobiology Laboratory, Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy.
| | | | | | | | | | | | | |
Collapse
|
25
|
Gründker C, Föst C, Fister S, Nolte N, Günthert AR, Emons G. Gonadotropin-releasing hormone type II antagonist induces apoptosis in MCF-7 and triple-negative MDA-MB-231 human breast cancer cells in vitro and in vivo. Breast Cancer Res 2010; 12:R49. [PMID: 20630060 PMCID: PMC2949636 DOI: 10.1186/bcr2606] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Revised: 05/12/2010] [Accepted: 07/14/2010] [Indexed: 12/31/2022] Open
Abstract
Introduction Triple-negative breast cancer does not express estrogen and progesterone receptors, and no overexpression/amplification of the HER2-neu gene occurs. Therefore, this subtype of breast cancer lacks the benefits of specific therapies that target these receptors. Today chemotherapy is the only systematic therapy for patients with triple-negative breast cancer. About 50% to 64% of human breast cancers express receptors for gonadotropin-releasing hormone (GnRH), which might be used as a target. New targeted therapies are warranted. Recently, we showed that antagonists of gonadotropin-releasing hormone type II (GnRH-II) induce apoptosis in human endometrial and ovarian cancer cells in vitro and in vivo. This was mediated through activation of stress-induced mitogen-activated protein kinases (MAPKs) p38 and c-Jun N-terminal kinase (JNK), followed by activation of proapoptotic protein Bax, loss of mitochondrial membrane potential, and activation of caspase-3. In the present study, we analyzed whether GnRH-II antagonists induce apoptosis in MCF-7 and triple-negative MDA-MB-231 human breast cancer cells that express GnRH receptors. In addition, we ascertained whether knockdown of GnRH-I receptor expression affects GnRH-II antagonist-induced apoptosis and apoptotic signaling. Methods Induction of apoptosis was analyzed by measurement of the loss of mitochondrial membrane potential. Apoptotic signaling was measured with quantification of activated MAPK p38 and caspase-3 by using the Western blot technique. GnRH-I receptor protein expression was inhibited by using the antisense knockdown technique. In vivo experiments were performed by using nude mice bearing xenografted human breast tumors. Results We showed that treatment of MCF-7 and triple-negative MDA-MB-231 human breast cancer cells with a GnRH-II antagonist results in apoptotic cell death in vitro via activation of stress-activated MAPK p38 and loss of mitochondrial membrane potential. In addition, we showed GnRH-II antagonist-induced activation of caspase-3 in MDA-MB-231 human breast cancer cells. After knockdown of GnRH-I receptor expression, GnRH-II antagonist-induced apoptosis and apoptotic signaling was only slightly reduced, indicating that an additional pathway mediating the effects of GnRH-II antagonists may exist. The GnRH-I receptor seems not to be the only target of GnRH-II antagonists. The antitumor effects of the GnRH-II antagonist could be confirmed in nude mice. The GnRH-II antagonist inhibited the growth of xenotransplants of human breast cancers in nude mice completely, without any apparent side effects. Conclusions GnRH-II antagonists seem to be suitable drugs for an efficacious and less-toxic endocrine therapy for breast cancers, including triple-negative breast cancers.
Collapse
Affiliation(s)
- Carsten Gründker
- Department of Gynecology and Obstetrics, Georg-August-University, Robert-Koch-Street 40, 37075 Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|
26
|
Pinto F, Calamo A, Totaro A, Sacco E, Volpe A, Racioppi M, D'Addessi A, Bassi P. Androgen-Deprivation Therapy in Prostate Cancer: Clinical Evidence and Future Perspectives. Urologia 2010. [DOI: 10.1177/039156031007700201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Androgens are involved in the development and progression of prostate cancer even if the mechanism is not well-recognized. For this reason androgen-deprivation therapy remains a milestone for the treatment of patients with advanced and metastatic disease and, in the last years, in conjunction with radiotherapy and surgery in locally advanced tumors. Alternative options, such as intermittent deprivation suppression, seem to be promising in terms of clinical benefits and toxicity profile. However, current therapies present side effects, such as testosterone surge with consequent clinical flare-up, metabolic syndrome and hormone-resistance, which develops after a variable number of years. Novel therapies such as LH-RH antagonists and prolonged depot LH-RH analogues have been developed in order to avoid clinical flare-up and testosterone microsurges. Novel androgen synthesis inhibitors, such as abiraterone acetate and MDV3100, have been recently discovered and tested as promising hormonal second-line agents in patients with castration-resistant prostate cancer. Finally, long-term side effects from androgen deprivation, such as osteoporosis, sarcopenic obesity and cardiovascular morbidity should be carefully monitored and properly treated.
Collapse
Affiliation(s)
- F. Pinto
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - A. Calamo
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - A. Totaro
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - E. Sacco
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - A. Volpe
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - M. Racioppi
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - A. D'Addessi
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| | - P.F. Bassi
- Urologia, Policlinico Universitario Agostino Gemelli, Università Cattolica del Sacro Cuore, Roma
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Fister S, Günthert AR, Aicher B, Paulini KW, Emons G, Gründker C. GnRH-II Antagonists Induce Apoptosis in Human Endometrial, Ovarian, and Breast Cancer Cells via Activation of Stress-Induced MAPKs p38 and JNK and Proapoptotic Protein Bax. Cancer Res 2009; 69:6473-81. [DOI: 10.1158/0008-5472.can-08-4657] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Clementi M, Sánchez C, Benitez DA, Contreras HR, Huidobro C, Cabezas J, Acevedo C, Castellón EA. Gonadotropin releasing hormone analogs induce apoptosis by extrinsic pathway involving p53 phosphorylation in primary cell cultures of human prostatic adenocarcinomas. Prostate 2009; 69:1025-33. [PMID: 19301301 DOI: 10.1002/pros.20954] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND Gonadotropin-releasing-hormone (GnRH) analogs are widely used to block hypothalamic-pituitary-gonadal axis and inhibit blood androgen levels in patients with prostate cancer (PCa). In addition, GnRH analogs induce proliferation arrest and apoptosis through GnRH receptors expressed on the membrane of PCa cells. Possible molecular mechanisms involved in GnRH-mediated apoptosis on prostate cancer cells were studied. METHODS Primary cultures from PCa and benign prostatic hyperplasia (BPH) (non-malignant control) were derived from samples provided by our Institutional Hospital. Cell cultures were incubated for 24 hr with 20 ng/ml of GnRH agonist Leuprolide (Lp) or antagonist Cetrorelix (Cx). Apoptosis was evaluated by studying the expression of Bax and Bcl-2 and the activation of caspase-9 (intrinsic pathway), caspase-8 (extrinsic pathway), and caspase-3. Also, mRNA level, protein expression and phosphorylation of p53 were studied. RESULTS Cleaved caspase-8 and -3, but not -9, increased in presence of Lp and Cx in PCa cell cultures. Bax and Bcl-2 mRNA levels showed no changes after GnRH-analog treatments. Only Bax protein showed an increase after Cx treatment in PCa cell cultures. p53 mRNA level was higher in PCa than in BPH cell cultures. Lp and Cx increased p53 expression and phosphorylation in PCa cell cultures. CONCLUSIONS Apoptosis induced by GnRH analogs seems to be mediated by extrinsic pathway involving p53 phosphorylation. Phosphorylated-p53 might be associated with the increase in apoptotic NGF receptor, p75, previously reported by our laboratory. These findings reinforce the concept of clinical use of GnRH analogs for PCa suggesting that intraprostatic treatment may be more effective.
Collapse
Affiliation(s)
- Marisa Clementi
- Faculty of Medicine, Physiology and Biophysics Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Signaling by G-protein-coupled receptor (GPCR): studies on the GnRH receptor. Front Neuroendocrinol 2009; 30:10-29. [PMID: 18708085 DOI: 10.1016/j.yfrne.2008.07.001] [Citation(s) in RCA: 213] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2007] [Revised: 04/28/2008] [Accepted: 07/21/2008] [Indexed: 01/22/2023]
Abstract
Gonadotropin-releasing hormone (GnRH) is the first key hormone of reproduction. GnRH analogs are extensively used in in vitro fertilization, and treatment of sex hormone-dependent cancers, due to their ability to bring about 'chemical castration'. The interaction of GnRH with its cognate type I receptor (GnRHR) in pituitary gonadotropes results in the activation of Gq/G(11), phospholipase Cbeta (PLCbetaI), PLA(2), and PLD. Sequential activation of the phospholipases generates the second messengers inositol 1, 4, 5-trisphosphate (IP(3)), diacylglycerol (DAG), and arachidonic acid (AA), which are required for Ca(2+) mobilization, the activation of various protein kinase C isoforms (PKCs), and the production of prostaglandin (PG) and other metabolites of AA, respectively. PKC isoforms are the major mediators of the downstream activation of a number of mitogen-activated protein kinase (MAPK) cascades by GnRH, namely: extracellular signal-regulated kinase (ERK), jun-N-terminal kinase (JNK), and p38MAPK. The activated MAPKs phosphorylate both cytosolic and nuclear proteins to initiate the transcriptional activation of the gonadotropin subunit genes and the GnRHR. While Ca(2+) mobilization has been found to initiate rapid gonadotropin secretion, Ca(2+), together with various PKC isoforms, MAPKs and AA metabolites also serve as key nodes, in the GnRH-stimulated signaling network that enables the gonadotropes to decode GnRH pulse frequencies and translating that into differential gonadotropin synthesis and release. Even though pulsatility of GnRH is recognized as a major determinant for differential gonadotropin subunit gene expression and gonadotropin secretion very little is yet known about the signaling circuits governing GnRH action at the 'Systems Biology' level. Direct apoptotic and metastatic effects of GnRH analogs in gonadal steroid-dependent cancers expressing the GnRHR also seem to be mediated by the activation of the PKC/MAPK pathways. However, the mechanisms dictating life (pituitary) vs. death (cancer) decisions made by the same GnRHR remain elusive. Understanding these molecular mechanisms triggered by the GnRHR through biochemical and 'Systems Biology' approaches would provide the basis for the construction of the dynamic connectivity maps, which operate in the various cell types (endocrine, cancer, and immune system) targeted by GnRH. The connectivity maps will open a new vista for exploring the direct effects of GnRH analogs in tumors and the design of novel combined therapies for fertility control, reproductive disorders and cancers.
Collapse
|
32
|
White CD, Coetsee M, Morgan K, Flanagan CA, Millar RP, Lu ZL. A crucial role for Galphaq/11, but not Galphai/o or Galphas, in gonadotropin-releasing hormone receptor-mediated cell growth inhibition. Mol Endocrinol 2008; 22:2520-30. [PMID: 18801931 DOI: 10.1210/me.2008-0122] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH acts on its cognate receptor in pituitary gonadotropes to regulate the biosynthesis and secretion of gonadotropins. It may also have direct extrapituitary actions, including inhibition of cell growth in reproductive malignancies, in which GnRH activation of the MAPK cascades is thought to play a pivotal role. In extrapituitary tissues, GnRH receptor signaling has been postulated to involve coupling of the receptor to different G proteins. We examined the ability of the GnRH receptor to couple directly to Galpha(q/11), Galpha(i/o), and Galpha(s), their roles in the activation of the MAPK cascades, and the subsequent cellular effects. We show that in Galpha(q/11)-negative cells stably expressing the GnRH receptor, GnRH did not induce activation of ERK, jun-N-terminal kinase, or P38 MAPK. In contrast to Galpha(i) or chimeric Galpha(qi5), transfection of Galpha(q) cDNA enabled GnRH to induce phosphorylation of ERK, jun-N-terminal kinase, and P38. Furthermore, no GnRH-mediated cAMP response or inhibition of isoproterenol-induced cAMP accumulation was observed. In another cellular background, [35S]GTPgammaS binding assays confirmed that the GnRH receptor was unable to directly couple to Galpha(i) but could directly interact with Galpha(q/11). Interestingly, GnRH stimulated a marked reduction in cell growth only in cells expressing Galpha(q), and this inhibition could be significantly rescued by blocking ERK activation. We therefore provide direct evidence, in multiple cellular backgrounds, that coupling of the GnRH receptor to Galpha(q/11), but not to Galpha(i/o) or Galpha(s), and consequent activation of ERK plays a crucial role in GnRH-mediated cell death.
Collapse
Affiliation(s)
- Colin D White
- The Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | | | | | | | | | | |
Collapse
|
33
|
Finch AR, Sedgley KR, Caunt CJ, McArdle CA. Plasma membrane expression of GnRH receptors: regulation by antagonists in breast, prostate, and gonadotrope cell lines. J Endocrinol 2008; 196:353-67. [PMID: 18252959 PMCID: PMC2229628 DOI: 10.1677/joe-07-0471] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In heterologous expression systems, human GnRH receptors (hGnRHRs) are poorly expressed at the cell surface and this may reflect inefficient exit from the endoplasmic reticulum. Here, we have defined the proportion of GnRHRs at the cell surface using a novel assay based on adenoviral transduction with epitope-tagged GnRHRs followed by staining and semi-automated imaging. We find that in MCF7 (breast cancer) cells, the proportional cell surface expression (PCSE) of hGnRHRs is remarkably low (<1%), when compared with Xenopus laevis (X) GnRHRs ( approximately 40%). This distinction is retained at comparable whole cell expression levels, and the hGnRHR PCSE is increased by addition of the XGnRHR C-tail (h.XGnRHR) or by a membrane-permeant pharmacological chaperone (IN3). The IN3 effect is concentration- and time-dependent and IN3 also enhances the hGnRHR-mediated (but not h.XGnRHR- or mouse GnRHR-mediated) stimulation of [(3)H]inositol phosphate accumulation and the hGnRHR-mediated reduction in cell number. We also find that the PCSE for hGnRHRs and h.XGnRHRs is low and is greatly increased by IN3 in two hormone-dependent cancer lines, but is higher and less sensitive to IN3 in a gonadotrope line. Finally, we show that the effect of IN3 on hGnRHR PCSE is not mimicked or blocked by two peptide antagonists although they do increase the PCSE for h.XGnRHRs, revealing that an antagonist-occupied cell surface GnRHR conformation can differ from that of the unoccupied receptor. The low PCSE of hGnRHRs and this novel peptide antagonist effect may be important for understanding GnRHR function in extrapituitary sites.
Collapse
|
34
|
Stangelberger A, Schally AV, Zarandi M, Heinrich E, Groot K, Havt A, Kanashiro CA, Varga JL, Halmos G. The combination of antagonists of LHRH with antagonists of GHRH improves inhibition of androgen sensitive MDA-PCa-2b and LuCaP-35 prostate cancers. Prostate 2007; 67:1339-53. [PMID: 17624923 DOI: 10.1002/pros.20605] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Antagonists of growth hormone-releasing hormone (GHRH) could extend the duration of response of androgen sensitive prostate cancers to androgen deprivation. METHODS We investigated the effect of new GHRH antagonists MZ-J-7-118 and MZ-J-7-138 and luteinizing hormone-releasing hormone (LHRH) antagonist Cetrorelix or castration on androgen sensitive MDA-PCa-2b and LuCaP-35 prostate cancer models xenografted into nude mice. Animals bearing androgen-independent LuCaP-35V prostatic cancer model were also treated with MZ-J-7-118. RESULTS Receptors for LHRH and GHRH were present in MDA-PCA-2b, LuCaP-35, and LuCaP-35V tumors. GHRH antagonists increased the inhibitory effect of surgical castration and LHRH antagonists on androgen sensitive MDA-PCa-2b and LuCaP-35 tumors. The time to relapse of androgen-dependent LuCaP-35 tumors was extended by GHRH antagonists. Growth of androgen-independent LuCaP-35V xenografts was also significantly inhibited by MZ-J-7-118. In MDA-PCa-2b tumors treatment with MZ-J-7-118 caused a significant decrease of VEGF and Cetrorelix or its combination with MZ-J-7-118 reduced EGF. The B(max) of EGF receptors was significantly reduced by Cetrorelix, MZ-J-7-118 and their combination. CONCLUSIONS Our findings suggest that the use of a combination of antagonists of GHRH and LHRH could improve the therapy for androgen sensitive prostate cancer. Antagonists of GHRH could be also considered for treatment of androgen-independent prostate cancers.
Collapse
Affiliation(s)
- Anton Stangelberger
- Veterans Affairs Medical Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Ovarian carcinoma continues to be the leading cause of death due to gynecological malignancy. Epidemiologic studies indicate that steroid hormones play roles in ovarian carcinogenesis. Gonadotropins, estrogen, and androgen may be causative factors, while gonadotropin-releasing hormone and progesterone may be protective factors in ovarian cancer pathogenesis. Experimental studies have shown that hormonal receptors are expressed in ovarian cancer cells and mediate the growth-stimulatory or growth-inhibitory effects of the hormones on these cells. Hormonal therapeutic agents have been evaluated in several clinical trials. Most of these trials were conducted in patients with recurrent or refractory ovarian cancer, with modest efficacy and few side effects. Better understanding of the mechanisms through which hormones affect cell growth may improve the efficacy of hormonal therapy. Molecular markers that can reliably predict major clinical outcomes should be investigated further in well-designed trials.
Collapse
Affiliation(s)
- H Zheng
- Department of Gynecologic Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | | | | | |
Collapse
|
36
|
Szabó J, Végh A, Rácz G, Szende B. Immunohistochemical demonstration of gonadotropin-releasing hormone receptors in prostate carcinoma. Urol Oncol 2006; 23:399-401. [PMID: 16301116 DOI: 10.1016/j.urolonc.2005.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2004] [Accepted: 03/03/2005] [Indexed: 10/25/2022]
Abstract
The antiandrogen and gonadotropin-releasing hormone (Gn-RH) analogue treatment of prostate carcinoma is based on decreased proliferative and increased apoptotic activity of tumor cells, induced by androgen ablation. Gn-RH analogues decrease the serum level of androgens by breaking the pituitary-gonadal axis, but increasing evidence points to the direct effect of Gn-RH analogues on tumor cells. Immunohistochemical demonstration of Gn-RH receptors recently became possible. Cryostat and paraffin sections of prostate biopsy samples of 10 untreated patients with prostate carcinoma (T2-T3, Gleason score 5-8) were investigated for Gn-RH receptors (Gn-RHR) and androgen receptors, respectively, using the immunoperoxidase method. Membrane bound, focal Gn-RHR positivity was found in 5 samples. Nuclear androgen receptor positivity appeared in 7 samples. No correlation between Gn-RHR and androgen receptor positivity was found, neither receptor status and tumor-nodes-metastasis stage nor Gleason score could be related to each other. Correlation between Gn-RHR positivity and response to luteinizing hormone-releasing hormone analogue treatment will be investigated further.
Collapse
Affiliation(s)
- Janos Szabó
- Department of Urology, Central Hospital of the Hungarian Army, Budapest, Hungary
| | | | | | | |
Collapse
|
37
|
Hasan J, Ton N, Mullamitha S, Clamp A, McNeilly A, Marshall E, Jayson GC. Phase II trial of tamoxifen and goserelin in recurrent epithelial ovarian cancer. Br J Cancer 2005; 93:647-51. [PMID: 16222310 PMCID: PMC2361624 DOI: 10.1038/sj.bjc.6602752] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Endocrine therapy is a recognised option in the treatment of chemo-resistant ovarian cancer. We conducted a nonrandomised phase II evaluation of combination endocrine therapy with tamoxifen and goserelin in patients with advanced ovarian cancer that had recurred following chemotherapy. In total, 26 patients entered the study, of which 17 had platinum-resistant disease. The median age was 63 years and enrolled patients had received a median of three chemotherapy regimens prior to trial entry. Patients were given oral tamoxifen 20 mg twice daily on a continuous basis and subcutaneous goserelin 3.6 mg once a month until disease progression. Using the definition of endocrine response that included patients with stable disease (SD) of 6 months or greater, the overall response rate (clinical benefit rate) was 50%. This included one complete response (CR) (3.8%), two partial responses (PR) (7.7%) and 10 patients with SD (38.5%). The median progression-free interval (PFI) was 4 months (95% CI 2.4–9.6) while the median overall survival (OS) was 13.6 months (95% CI 5.5–30.6). Four patients received treatment for more than 2 years (range 1–31) and one of them is still on treatment. In none of the four patients was there any evidence of recurrent or cumulative treatment related toxicity. Treatment-limiting toxicity was not seen in any of the study population. Endocrine data demonstrated a marked suppression of luteinising hormone (LH) and follicle-stimulating hormone (FSH) to less than 4% of baseline values. No consistent correlation could be established between LH/FSH suppression and tumour response. Likewise no relationship was observed between Inhibin A/B and pro-alpha C levels and tumour response. Inhibin is unlikely to be a useful surrogate marker for response in locally advanced or metastatic ovarian cancer. Combination endocrine therapy with tamoxifen and goserelin is an active regimen in platinum-resistant ovarian cancer patients. Hormonal therapy is advantageous in its relative lack of toxicity, ease of administration and tolerability, thus making it suitable for patients with heavily pretreated disease, compromised bone marrow function and other comorbid conditions that contraindicate cytotoxic therapy as well as in patients with indolent disease.
Collapse
Affiliation(s)
- J Hasan
- Cancer Research UK, Department Medical Oncology, Christie Hospital, Wilmslow Road, Withington, Manchester M20 4BX, UK.
| | | | | | | | | | | | | |
Collapse
|
38
|
Rispoli LA, Nett TM. Pituitary gonadotropin-releasing hormone (GnRH) receptor: structure, distribution and regulation of expression. Anim Reprod Sci 2005; 88:57-74. [PMID: 15993012 DOI: 10.1016/j.anireprosci.2005.05.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Reproduction in mammals is controlled by interactions between the hypothalamus, anterior pituitary and gonads. Interaction of GnRH with its cognate receptor is essential to regulating reproduction. Characterization of the structure, distribution and expression of GnRH receptors (GnRH-R) has furthered our understanding of the physiological consequences of GnRH stimulation of pituitary gonadotropes. Based on the putative topology of the amino acid sequence of the GnRH-R and point mutation studies, key elements of the GnRH-R have been identified to play a role in ligand recognition and binding, G-protein activation and internalization. Normally, reproductive function is mediated by GnRH-R expressed only on the membranes of pituitary gonadotropes. The density of GnRH-R on gonadotropes determines their ability to respond to GnRH. This density is highest just prior to ovulation and likely is important for complete expression of the pre-ovulatory surge of LH. Therefore, knowledge regarding what regulates the density of GnRH-R is essential to understanding changes in pituitary sensitivity to GnRH and ultimately, to expression of the LH surge. Regulation of GnRH-R gene expression is influenced by a multitude of factors including gonadal steroid hormones, inhibin, activin and perhaps most importantly GnRH itself.
Collapse
Affiliation(s)
- L A Rispoli
- Department of Biomedical Sciences, Animal Reproduction and Biotechnology Laboratory, Colorado State University, Fort Collins, CO 80523, USA
| | | |
Collapse
|
39
|
Hapgood JP, Sadie H, van Biljon W, Ronacher K. Regulation of expression of mammalian gonadotrophin-releasing hormone receptor genes. J Neuroendocrinol 2005; 17:619-38. [PMID: 16159375 DOI: 10.1111/j.1365-2826.2005.01353.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH), acting via its cognate GnRH receptor (GnRHR), is the primary regulator of mammalian reproductive function, and hence GnRH analogues are extensively used in the treatment of hormone-dependent diseases, as well as for assisted reproductive techniques. In addition to its established endocrine role in gonadotrophin regulation in the pituitary, evidence is rapidly accumulating to support the expression and functional roles for two forms of GnRHR (GnRHR I and GnRHR II) in multiple and diverse extra-pituitary mammalian tissues and cells. These findings, together with findings indicating that mutations of the GnRHR are linked to the disease hypogonadotrophic hypogonadism and that GnRHRs play a direct role in neuronal migration and reproductive cancers, have presented new therapeutic targets and intensified research into the structure, function and mechanisms of regulation of expression of GnRHR genes. The present review focuses on the current knowledge on tissue-specific and hormonal regulation of transcription of mammalian GnRH receptor genes. Emerging insights, such as the discovery of diverse regulatory mechanisms in pituitary and extra-pituitary cell types, nonclassical mechanisms of steroid regulation, the use of composite elements for cell-specific expression, the increasing profile of hormones involved in regulation, the complexity of kinase pathways that target the GnRHR I gene, as well as species-differences, are highlighted. Although further research is necessary to understand the mechanisms of regulation of expression of GnRHR I and GnRHR II genes, the GnRHR is emerging as a potential target gene for facilitating cross-talk between neuroendocrine, immune and stress-response systems in multiple tissues via autocrine, paracrine and endocrine signalling.
Collapse
Affiliation(s)
- J P Hapgood
- Department of Biochemistry, University of Stellenbosch, Matieland, South Africa.
| | | | | | | |
Collapse
|
40
|
Bodek G, Vierre S, Rivero-Müller A, Huhtaniemi I, Ziecik AJ, Rahman NA. A novel targeted therapy of Leydig and granulosa cell tumors through the luteinizing hormone receptor using a hecate-chorionic gonadotropin beta conjugate in transgenic mice. Neoplasia 2005; 7:497-508. [PMID: 15967102 PMCID: PMC1501163 DOI: 10.1593/neo.04751] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Revised: 12/31/2004] [Accepted: 01/04/2005] [Indexed: 11/18/2022] Open
Abstract
We investigated the antitumoral efficacy, endocrine consequences, and molecular mechanisms underlying cell death induced by the Hecate-chorionic gonadotropin (CG)beta conjugate, a fusion protein of a 23-amino acid lytic peptide Hecate with a 15-amino acid (81-95) fragment of the human CGbeta chain. Transgenic (TG) mice expressing the inhibin alpha-subunit promoter (inhalpha)/Simian Virus 40 T-antigen (Tag) transgene, developing luteinizing hormone (LH) receptor (R) expressing Leydig and granulosa cell tumors, and wild-type control littermates were treated either with vehicle, Hecate, or Hecate-CGbeta conjugate for 3 weeks. Hecate-CGbeta conjugate treatment reduced the testicular and ovarian tumor burden (P < .05), whereas a concomitant increase (testis; P < .05) or no change (ovary) in tumor volumes occured with Hectate treatment. A drop in serum progesterone, produced by the tumors, and an increase in LH levels occured in Hecate-CGbeta treated mice, in comparison with vehicle and Hecate groups, providing further support for the positive treatment response. Hecate-CGbeta conjugate induced a rapid and cell-specific membrane permeabilization of LHR-expressing cells in vitro, suggesting a necrotic mode of cell death without activation of apoptosis. These results prove the principle that the Hecate-CGbeta conjugate provides a novel specific lead into gonadal somatic cell cancer therapy by targeted destruction of LHR-expressing tumor cells.
Collapse
MESH Headings
- Amino Acid Chloromethyl Ketones/pharmacology
- Animals
- Apoptosis
- Blotting, Northern
- Caspase 3
- Caspases/metabolism
- Cell Death
- Cell Line, Tumor
- Cell Separation
- Chorionic Gonadotropin, beta Subunit, Human/chemistry
- Chorionic Gonadotropin, beta Subunit, Human/therapeutic use
- Disease Models, Animal
- Enzyme Activation
- Female
- Flow Cytometry
- Granulosa Cell Tumor/therapy
- Humans
- Leydig Cell Tumor/therapy
- Male
- Melitten/analogs & derivatives
- Melitten/chemistry
- Melitten/therapeutic use
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Fluorescence
- Necrosis
- Ovarian Neoplasms/therapy
- Progesterone/blood
- Promoter Regions, Genetic
- Protein Structure, Tertiary
- Receptors, LH/metabolism
- Recombinant Fusion Proteins/metabolism
- Testicular Neoplasms/therapy
- Time Factors
Collapse
Affiliation(s)
- Gabriel Bodek
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn 10-714, Poland
| | | | | | | | | | | |
Collapse
|
41
|
Sorianello EM, Fernandez MO, Catalano PN, Mongiat LA, Somoza GM, Libertun C, Lux-Lantos VA. Differential gonadotropin releasing hormone (GnRH) expression, autoregulation and effects in two models of rat luteinized ovarian cells. Life Sci 2005; 77:2141-55. [PMID: 16005024 DOI: 10.1016/j.lfs.2005.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2005] [Accepted: 03/03/2005] [Indexed: 11/15/2022]
Abstract
GnRH has been suggested to participate in corpus luteum function. Here we studied the expression of GnRH mRNA and peptide in two models of rat luteinized tissues: ovarian cells from PMSG-hCG treated prepubertal rats (SPO) and from intrasplenic ovarian tumors (Luteoma). A GnRH autoregulatory effect was evaluated as well as its action on cell proliferation and apoptosis. GnRH mRNA was present in SPO, isolated corpora lutea from SPO and Luteoma from 1 week to 7 months of development. In vitro cultures of Luteoma cells expressed 2-fold higher GnRH mRNA and 10-fold higher GnRH peptide than SPO cells. Buserelin (GnRH analog) increased GnRH mRNA and peptide expression in SPO but not in Luteoma cells. While basal proliferation was very low in Luteoma cells, SPO cells showed a significant increase in cell number by both the thymidine and the MTS methods after 72 h in culture. Buserelin induced a decrease in cell number in both cell types to a similar degree. Although basal apoptosis levels were higher in SPO than in Luteoma cells, Buserelin-induced apoptosis was only detected in Luteoma cells after 48 h treatment. These results show that the two types of rat, luteinized tissues, Luteoma and SPO, markedly differed in some intrinsic properties and in their local GnRH systems. Luteoma cells proliferate very weakly, express and secrete high amounts of GnRH, do not show an autoregulatory effect and respond to the decapeptide with apoptosis stimulation. In contrast SPO cells proliferate significantly, secrete low levels of GnRH but possess a positive, autoregulatory mechanism and respond to GnRH stimulation with impairment of proliferation.
Collapse
Affiliation(s)
- E M Sorianello
- Neuroendocrinology Laboratory, Instituto de Biología y Medicina Experimental-CONICET, Vuelta de Obligado 2490, (C1428ADN) Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
42
|
Tieva A, Bergh A, Damber JE. Alteration of gonadotropin-releasing hormone receptor expression with the progression of prostate cancer in the Dunning rat adenocarcinoma sublines. Acta Oncol 2005; 44:299-303. [PMID: 16076703 DOI: 10.1080/02841860510007512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Inhibitory effects of GnRH analogues on tumour growth in vitro suggests that such direct effects may be of importance also in vivo. However, the role of GnRH receptors (GnRH-R) in prostate tumour progression is largely unknown. The aim was therefore to investigate the variation of GnRH-R expression with prostate tumour progression using Dunning rat adenocarcinoma sublines representing different prostate tumour grades. GnRH-R levels were quantified in the rat dorsolateral (DLP) and Dunning sublines (PAP, AT-1, AT-2, AT-3, MatLyLu) using competitive RT-PCR and Western blot. The results showed that all Dunning sublines had significantly elevated GnRH-R mRNA expression levels compared with DLP. Comparison of GnRH-R mRNA levels between different tumour grades revealed no difference in mRNA expression. However, the anaplastic and highly metastatic AT-3 and MatLyLu tumours displayed a tendency for lower GnRH-R mRNA values than the non-metastatic tumour sublines. Our data demonstrate the expression of GnRH-R in normal rat DLP and in different Dunning sublines. However, GnRH-R seems not to be involved in tumour progression.
Collapse
Affiliation(s)
- Ase Tieva
- Institute of Surgical and Perioperative Sciences, Urology & Andrology, Umeå University, Umeå, Sweden.
| | | | | |
Collapse
|
43
|
Malmusi S, La Marca A, Giulini S, Xella S, Tagliasacchi D, Marsella T, Volpe A. Comparison of a gonadotropin-releasing hormone (GnRH) antagonist and GnRH agonist flare-up regimen in poor responders undergoing ovarian stimulation. Fertil Steril 2005; 84:402-6. [PMID: 16084881 DOI: 10.1016/j.fertnstert.2005.01.139] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2004] [Revised: 01/31/2005] [Accepted: 01/31/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To compare the efficacy of flare-up and GnRH-antagonist treatment in poor-responder patients. DESIGN Randomized prospective study. SETTING Assisted reproduction center. PATIENT(S) Fifty-five poor-responder patients undergoing intracytoplasmic sperm injection (ICSI). INTERVENTION(S) Thirty patients received GnRH agonist on the 1st day of menstruation, followed by exogenous gonadotropins from the 2nd day. Twenty-five patients received exogenous gonadotropins starting on the second day of menstruation, followed by GnRH antagonist when the leading follicle reached 14 mm in diameter. MAIN OUTCOME MEASURE(S) The total dose of FSH administered during the ovarian stimulation, as well as the number of mature oocytes retrieved, embryo quality, fertilization, implantation, and pregnancy rates were evaluated. RESULT(S) The number of ampules and units of FSH administered were significantly less in the flare-up than in the antagonistic group. The numbers of mature oocytes retrieved and of top-quality embryos transferred were significantly greater in the flare-up than in the GnRH-antagonist group. The fertilization rate (84% vs. 63%) was significantly higher in the flare-up than in the GnRH-antagonist group. The implantation and pregnancy rate were similar in the two groups. CONCLUSION(S) The flare-up protocol appears to be more effective than the GnRH-antagonist protocol in terms of mature oocytes retrieved, fertilization rate, and top-quality embryos transferred in poor-responder patients.
Collapse
Affiliation(s)
- Stefania Malmusi
- Institute of Obstetrics and Gynecology, Policlinico of Modena, Modena University, Modena, Italy.
| | | | | | | | | | | | | |
Collapse
|
44
|
Nagy A, Schally AV. Targeting of cytotoxic luteinizing hormone-releasing hormone analogs to breast, ovarian, endometrial, and prostate cancers. Biol Reprod 2005; 73:851-9. [PMID: 16033997 DOI: 10.1095/biolreprod.105.043489] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Targeted chemotherapy is a modern approach aimed at increasing the efficacy of systemic chemotherapy and reducing its side effects. The peptide receptors expressed primarily on cancerous cells can serve as targets for a selective destruction of malignant tumors. Binding sites for LHRH (now known in genome and microarray databases as GNRH1), were found on 52% of human breast cancers, about 80% of human ovarian and endometrial cancers, and 86% of human prostatic carcinoma specimens. Because LHRH receptors are not expressed on most normal tissues, they represent a specific target for cancer chemotherapy with antineoplastic agents linked to an LHRH vector molecule. To test the efficacy of targeted chemotherapy based on LHRH analogs, we recently developed a cytotoxic analog of LHRH, designated AN-152, which consists of [D-Lys6]LHRH covalently linked to one of the most widely used chemotherapeutic agents, doxorubicin (DOX). In addition, we designed and synthesized a highly active derivative of DOX, 2-pyrrolino-DOX (AN-201), which is 500-1000 times more potent than DOX in vitro. AN-201 is active against tumors resistant to DOX, and noncardiotoxic. As in the case of DOX, AN-201 was coupled to carrier peptide [D-Lys6]LHRH to form a superactive targeted cytotoxic LHRH analog, AN-207. Both AN-152 and AN-207 can effectively inhibit the growth of LHRH receptor-positive human breast, ovarian, endometrial, and prostate cancers xenografted into nude mice. DOX-containing cytotoxic LHRH analog AN-152 is scheduled for clinical phase I/IIa trials in patients with advanced ovarian and breast cancers in 2005.
Collapse
Affiliation(s)
- Attila Nagy
- Section of Experimental Medicine, Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana 70112, USA
| | | |
Collapse
|
45
|
Cheng CK, Leung PCK. Molecular biology of gonadotropin-releasing hormone (GnRH)-I, GnRH-II, and their receptors in humans. Endocr Rev 2005; 26:283-306. [PMID: 15561800 DOI: 10.1210/er.2003-0039] [Citation(s) in RCA: 177] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
In human beings, two forms of GnRH, termed GnRH-I and GnRH-II, encoded by separate genes have been identified. Although these hormones share comparable cDNA and genomic structures, their tissue distribution and regulation of gene expression are significantly dissimilar. The actions of GnRH are mediated by the GnRH receptor, which belongs to a member of the rhodopsin-like G protein-coupled receptor superfamily. However, to date, only one conventional GnRH receptor subtype (type I GnRH receptor) uniquely lacking a carboxyl-terminal tail has been found in the human body. Studies on the transcriptional regulation of the human GnRH receptor gene have indicated that tissue-specific gene expression is mediated by differential promoter usage in various cell types. Functionally, there is growing evidence showing that both GnRH-I and GnRH-II are potentially important autocrine and/or paracrine regulators in some extrapituitary compartments. Recent cloning of a second GnRH receptor subtype (type II GnRH receptor) in nonhuman primates revealed that it is structurally and functionally distinct from the mammalian type I receptor. However, the human type II receptor gene homolog carries a frameshift and a premature stop codon, suggesting that a full-length type II receptor does not exist in humans.
Collapse
Affiliation(s)
- Chi Keung Cheng
- Department of Obstetrics and Gynecology, University of British Columbia, Vancouver, British Columbia, Canada V6H 3V5
| | | |
Collapse
|
46
|
Yates C, Wells A, Turner T. Luteinising hormone-releasing hormone analogue reverses the cell adhesion profile of EGFR overexpressing DU-145 human prostate carcinoma subline. Br J Cancer 2005; 92:366-75. [PMID: 15655536 PMCID: PMC2361841 DOI: 10.1038/sj.bjc.6602350] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cetrorelix, a luteinising hormone-releasing hormone (LHRH) analogue, has been shown to limit growth of the human androgen-independent prostate cell line DU-145, although other inhibitory actions may also be affected. Both growth and invasion of DU-145 cells are linked to autocrine epidermal growth factor receptor (EGFR) signalling. Invasiveness requires not only cells to migrate to conduits, but also reduced adhesiveness between tumour cells to enable separation from the tumour mass. Thus, we investigated whether Cetrorelix alters the DU-145 cell-cell adhesion and if this occurs via altered EGFR signalling. Pharmacologic levels of Cetrorelix limited the invasiveness of a highly invasive DU-145 subline overexpressing full-length EGFR (DU-145 WT). Extended exposure of the cells to Cetrorelix resulted in increased levels of the cell-cell adhesion complex molecules E-cadherin, alpha- and beta-catenin, and p120. Puromycin blocked the increases in E-cadherin and beta-catenin levels, suggesting that de novo protein synthesis is required. The Cetrorelix effect appears to occur via transmodulation of EGFR by a protein kinase C (PKC)-dependent mechanism, as there were no changes in DU-145 cells expressing EGFR engineered to negate the PKC transattenuation site (DU-145 A654); downregulation of EGFR signalling produced a similar upregulation in adhesion complex proteins, further suggesting a role for autocrine signalling. Cetrorelix increased the cell-cell adhesiveness of DU-145 WT cells to an extent similar to that seen when autocrine EGFR signalling is blocked; as expected, DU-145 A654 cell-cell adhesion also was unaffected by Cetrorelix. The increased adhesiveness is expected as the adhesion complex molecules moved to the cells' periphery. These data offer direct insight into the possible crosstalk pathways between the LHRH and EGFR receptor signalling. The ability of Cetrorelix to downregulate EGFR signalling and subsequently reverse the antiadhesiveness found in metastatic prostate cancer highlights a novel potential target for therapeutic strategies.
Collapse
Affiliation(s)
- C Yates
- Department of Pathology, University of Pittsburgh, and Pittsburgh VAMC, Pittsburgh, PA 15261, USA
- Department of Biology and Center for Cancer Research, Carver Research Foundation, Tuskegee University, Tuskegee, AL 36088, USA
| | - A Wells
- Department of Pathology, University of Pittsburgh, and Pittsburgh VAMC, Pittsburgh, PA 15261, USA
| | - T Turner
- Department of Biology and Center for Cancer Research, Carver Research Foundation, Tuskegee University, Tuskegee, AL 36088, USA
- Department of Biology and Center for Cancer Research, Carver Research Foundation, Tuskegee University, Tuskegee, AL 36088, USA. E-mail:
| |
Collapse
|
47
|
Sepp-Lorenzino L, Slovin S. Prostate cancer: therapeutic patent review. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.10.12.1833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Chen W, Yoshida S, Ohara N, Matsuo H, Morizane M, Maruo T. Gonadotropin-releasing hormone antagonist cetrorelix down-regulates proliferating cell nuclear antigen and epidermal growth factor expression and up-regulates apoptosis in association with enhanced poly(adenosine 5'-diphosphate-ribose) polymerase expression in cultured human leiomyoma cells. J Clin Endocrinol Metab 2005; 90:884-92. [PMID: 15536154 DOI: 10.1210/jc.2004-1591] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The objective of this study was to elucidate the effects of GnRH antagonist Cetrorelix on proliferation and apoptosis in human leiomyoma cells cultured in vitro. Isolated leiomyoma cells were subcultured in phenol red-free DMEM supplemented with 10% fetal bovine serum for 120 h and then stepped down to serum-free conditions in the presence or absence of graded concentrations of Cetrorelix (10(-5) to 10(-8) mol/liter) for 6 d. Cultured leiomyoma cells were used for semiquantitative RT-PCR, immunocytochemistry, Western blot analysis, and terminal deoxynucleotidyl transferase-mediated deoxyuridine 5-triphosphate nick-end labeling assay. RT-PCR analysis revealed the presence of mRNAs encoding for GnRH receptor and epidermal growth factor (EGF) in cultured leiomyoma cells. The number of viable cultured leiomyoma cells was significantly (P < 0.01) decreased by treatment with Cetrorelix compared with untreated control cultures. Immunocytochemical examination demonstrated that treatment with Cetrorelix attenuated the expression of proliferating cell nuclear antigen (PCNA) and EGF in cultured leiomyoma cells. Western blot analysis revealed that treatment with 10(-5) mol/liter Cetrorelix significantly (P < 0.01) decreased PCNA expression. In addition, treatment with 10(-5) mol/liter Cetrorelix remarkably increased the terminal deoxynucleotidyl transferase-mediated deoxyuridine 5-triphosphate nick-end labeling-positive rate and poly(ADP-ribose) polymerase expression at 24 h of treatment compared with untreated control cultures (P < 0.01). Furthermore, treatment with 10(-5) mol/liter Cetrorelix decreased immunoreactive EGF protein and EGF mRNA expression in cultured leiomyoma cells at 4 d of treatment. GnRH antagonist Cetrorelix may directly inhibit leiomyoma cell growth by down-regulating proliferation in association with a decrease in EGF mRNA expression and by up-regulating apoptosis in those cells.
Collapse
Affiliation(s)
- Wei Chen
- Department of Obstetrics and Gynecology, Kobe University Graduate School of Medicine, Chuo-Ku, Kobe 650-0017, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Gnanapragasam VJ, Darby S, Khan MM, Lock WG, Robson CN, Leung HY. Evidence that prostate gonadotropin-releasing hormone receptors mediate an anti-tumourigenic response to analogue therapy in hormone refractory prostate cancer. J Pathol 2005; 206:205-13. [PMID: 15818594 DOI: 10.1002/path.1767] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Gonadotropin-releasing hormone analogue (GnRHa) therapy is an established method of androgen withdrawal in the treatment of prostate cancer. The present study investigated if the expression of prostate GnRH receptors (GnRHRs) might influence the response to GnRHa. GnRHR protein expression was first studied in a panel of prostate cancer cell lines. In androgen-dependent cells, GnRHR expression was unchanged following acute or chronic androgen withdrawal. In these cells, GnRHa significantly inhibited androgen-induced cell proliferation (p = 0.01). In contrast, GnRHa was unable to further suppress basal levels of cell proliferation induced by androgen withdrawal. In androgen-independent prostate cancer cells, variable levels of GnRHR expression were observed. In these cells, GnRHa treatment blocked cell proliferation (p = 0.001) and invasion (up to 70%) induced by fibroblast growth factor stimulation. Crucially, this effect was only evident in cells that expressed high levels of the GnRHR. GnRHa treatment also significantly inhibited the ability of these cells to recover from a cytotoxic insult (50% inhibition). The clinical significance of prostate GnRHR was tested by immunohistochemistry in a preliminary cohort of patients treated with GnRHa or surgical castration. There was no association between GnRHR expression and pathological grade, clinical stage, time to PSA nadir (p = 0.82) (n = 35) or progression to hormone refractory disease (p = 0.22) (n = 21), irrespective of the treatment method. GnRHa therapy in the presence of high GnRHR expression however, was found to be associated with longer disease-specific survival (mean survival 85 months, p = 0.002). In contrast, high GnRHR expression was not associated with survival among surgically castrated patients (mean survival 50 months, p = 0.7). Taken together, these data support the notion of a functional interaction between GnRHa and the GnRHR, which results in an anti-tumourigenic effect on prostate cancer cells. Findings from this report have direct implications for the use of GnRHR as a novel therapeutic target in hormone refractory prostate cancer.
Collapse
Affiliation(s)
- V J Gnanapragasam
- Urology Research Group, Northern Institute for Cancer Research, Medical School, University of Newcastle upon Tyne, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Hypothalamic gonadotropin releasing hormone (GnRH I) and its pituitary receptor are responsible for the CNS regulation of reproduction. However, a second GnRH (GnRH II) is also expressed in humans and a gene that resembles the GnRH II receptor in fish has been identified in humans and monkeys. The amino-acid sequence of this newly identified, seven-transmembrane, G-protein-coupled receptor in monkeys differs from the human GnRH I receptor by having a C-terminal, cytoplasmic tail. GnRH II is approximately 400-fold more potent at GnRH II receptors than GnRH I receptors. GnRH I directly inhibits proliferation of human tumor cells, and GnRH II and its receptor might have a similar role. Limited progress has been made, however, because of difficulty translating the mRNA that encodes the human GnRH II receptor. Nevertheless, such receptors are likely to exist in humans because GnRH II is more inhibitory to tumor cell replication than GnRH I, and GnRH I and GnRH II have reciprocal effects on human decidual stromal cells in culture. The focus of this review is the identity of a possible translatable, functional GnRH II receptor in humans. The two possibilities considered are either that GnRH II receptor mRNA is expressed that encodes either 5 or 7 transmembrane domains or that a GnRH II-responsive complex is formed by the GnRH I receptor and fragments derived from the GnRH II receptor.
Collapse
Affiliation(s)
- Jimmy D Neill
- University of Alabama School of Medicine, Department of Physiology and Biophysics, MCLM 816, 1918 University Boulevard, Birmingham, AL 35294-0005, USA.
| | | | | |
Collapse
|