The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review)

  • Authors:
    • Arturo Aguilar-Rojas
    • Marco Allan Pérez-Solis
    • Guadalupe Maya-Núñez
  • View Affiliations

  • Published online on: January 19, 2016     https://doi.org/10.3892/ijo.2016.3346
  • Pages: 861-868
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recently, an increasing amount of evidence indicates that human gonadotropin-releasing hormone (hGnRH) and its receptor (hGnRHR) are important regulatory components not only to the reproduction process but also in the regulation of some cancer cell functions such as cell proliferation, in both hormone-dependent and -independent types of tumors. The hGnRHR is a naturally misfolded protein that is retained mostly in the endoplasmic reticulum; however, this mechanism can be overcome by treatment with several pharmacoperones, therefore, increasing the amount of receptors in the cell membrane. In addition, several reports indicate that the expression level of hGnRHR in tumor cells is even lower than in pituitary or gonadotrope cells. The signal transduction pathways activated by hGnRH in both gonadotrope and different cancer cell types are described in the present review. We also discuss how the rescue of misfolded receptors in tumor cells could be a promising strategy for cancer therapy.

1. Introduction

Cancer remains among the leading causes of mortality word-wide with an increase of 7.5 million of deaths in 2008 to 8.2 million in 2012. Furthermore, 32.6 million people are living with cancer according to the latest GLOBOCAN estimated cancer incidence, mortality and prevalence (http://globocan.iarc.fr/Pages/fact_sheets_cancer.aspx). Despite the advances in the diagnosis and treatment of human malignancy, it is necessary to find new improved strategies to treat cancer. The human gonadotropin-releasing hormone (hGnRH) is a key hormone in the regulation of reproduction. However, an increasing number of reports have been shown the participation of GnRH and its receptor, not only in the reproduction but also in the regulation of tumor cell behavior. The hGnRH has been shown effective in controlling cell growth and invasiveness in certain type of tumors, both in vivo and in vitro. Furthermore, in some cases it has been reported to possess anti-oncogenic activity. These characteristics make the hGnRH/hGnRH receptor (hGnRHR) an ideal model in the study of new approaches for cancer treatment. The present review will focus on the signal transduction pathways activated when the hormone binds its receptor, in gonadotrope cells and in several types of cancer and how the relatively new concept of hGnRHR overexpression by pharmacoperons could be a new therapeutic strategy for cancer treatment.

2. GnRH receptor signaling in the pituitary

The tissue distribution pattern of the hGnRH and its receptor is wide and diversified. Therefore, it is not surprising that the cell signaling pathways produced from the interaction of the receptor with its hormone largely depend on the cellular context in which this occurs (1). In pituitary gonadotropes, the interaction between hGnRHR with its ligand induces conformational changes in both, the receptor itself and the coupled G-protein. The G protein family belongs to the heterotrimeric GTPases that are integrated by three subunits: Gα, Gβ and Gγ. There is a wide range of G proteins, which have been subclassified based on their structural differences in the Gα subunit. However, in gonadotrope cells the hGnRHR can be coupled to Gαq/11, and according to some reports, to Gαs subunit (2). The structural change of the G protein modifies the affinity for GDP, and the guanine nucleotide exchange factor facilitates the passive substitution of GDP for GTP. This nucleotide exchange causes the Gα subunit separation of the heterotrimer without affecting the association between the Gβ and Gγ subunits, which remain as one (3). Early studies on hGnRH signaling pathway in gonadotrophs, showed that only the Gαq/11 subunit could initiate an intracellular signaling cascade, through its main effector, the phospholipase Cβ (PLCβ) (4). However, further functional studies have shown that Gβγ dimer is also able to activate PLCβ; in fact it has been observed that in response to a sustained stimulation of hGnRH other effectors such as phospholipase A2 (PLA2) and D (PLD) can also be activated consecutively through both, Gα or Gβγ (Fig. 1) (5).

Figure 1

Signal transduction pathways activated by hGnRH. In pituitary cells (left panel) the hGnRHR once activated by its ligand is able to couple to Gαq/11 protein initiating the signal transduction pathway mediated by protein kinase C (PKC). This kinase is able to phosphorylate and activate another protein the phospholipase D (PLD) and the latest also phosphorylate the fibrosarcoma protein kinase 1 (Raf-1), protein tyrosine kinase SRC and some mitogen-activated protein kinases (MAPKs). The activation of Raf-1/MAPKs ends with the phosphorylation of transcription factors, such as Elk-1, c-Fos and c-Jun, which positively regulate the transcription of the gonadotropin α subunit gene, of the same PKC gene and the phospholipase A2 gene (PLA2). On the other hand in cancer cells (right panel) the receptor once activated by GnRH is able to couple both Gαi and Gαq/11 proteins depending on the cell background. When the receptor couples to Gαi it activates the adenylate cyclase (AC) that converts ATP in cyclic AMP (cAMP) that also in turn activate the protein kinase A (PKA). Once the PKA signal tranduction pathway is active the kinase phosphorylates and activated downstream kinases that directly or indirectly induce a decrease in epidermal growth factor receptor (EGFR), insulin-like growth factor I receptor (IGF-1R) and insulin-like growth factor 1 (IGF-1) that inhibit cell proliferation. In ovarian cancer, for instance, the hGnRHR also couples to Gαq/11 protein initiating the PKC signaling pathway resulting also in inhibition of cell proliferation.

Phospholipase C uses phosphatidylinositol 4,5 bisphosphate (PIP2) as substrate and the enzymatic hydrolysis of this phospholipid produces the first wave of the second messengers, diacylglycerol (DAG) and inositol 1,4,5 triphosphate (IP3) (2). The endoplasmic reticulum membrane contains IP3 receptors that can function as Ca2+ channels when activated by their specific ligand. The interaction of IP3 changes the structural conformation of its receptor, causing an increase in Ca2+ channel sensitivity with the consequent release of the cation into the cytosol. Subsequently, an increase in the concentration of Ca2+ activates the L-type voltage-sensitive calcium channels located in the cell membrane (6,7). Several studies have demonstrated the functional association between the accumulation of intracellular Ca2+ induced by hGnRH and the exocytosis of gonadotropins (8). In gonadotrope cells, the production of DAG and Ca2+ release into the cytosol are the main events in the synthesis and release of gonadotropins in response to hGnRH through the activation of protein kinase C (PKC) (9).

Due to the pleiotropic activity of the PKC, its activation is considered to be the central event in the activation of the hGnRH system. Studies performed in different cell systems have reported that two PKC isoforms α and βII have the ability to directly phosphorylate several residues of PLD and therefore activate the kinase (10,11). PLD hydrolyzes the phosphatidylcholine (PC) producing fosfatidilethanol (PET) and phosphatidic acid (PA), and both can act as second messengers increasing furthermore the signaling pathways initiated by hGnRH. PLD also rapidly activates fibrosarcoma protein kinase 1 (Raf-1), protein tyrosine kinase SRC and some mitogen-activated protein kinases (MAPKs) that are the immediate effectors in gonadotropes (10,1214).

The direct or indirect activation of PKC upon via the Raf-1/MAPKs ends with the phosphorylation of transcription factors, such as Elk-1, c-Fos and c-Jun, which positively regulate the transcription of the gonadotropin α subunit gene and the phospholipase A2 (PLA2) gene (2,15,16). In the gonadotrope cell line αT3-1, the PLA2 generates arachidonic acid (AA), which in turn is substrate for the lipoxygenase that converts AA into leukotrienes. These leukotrienes are also involved in the induction of gene expression of the gonadotropins α subunit as well as PKCβ (17,18). In turn all the signal transduction pathways induced by hGnRH and transduced by the axis hGnRHR/Gαq/11/PLC/PKC, culminate in the event of synthesis and release of gonadotropins, luteinizing hormone (LH) and follicle-stimulating hormone (FSH) (Fig. 1) (19). The activation of a second G protein by the hGnRH stimulation in the gonadotrope remains controversial, some reports have indicated the activation of Gαs with the consequent activation of adenylate cyclase (AC) and the formation of the second messenger, cyclic adenosine monophosphate (cAMP), which in turn is the cofactor of the protein kinase A (PKA); however, further experiments are necessary to confirm these results (20).

Another component of these complex networks of signal transduction pathways is regulated by both frequency and amplitude of pulses of hypothalamic hGnRH secretion into the bloodstream depending on the physiological needs of the organism (21). It has been reported that when gonadotrope cells are exposed to a high frequency pulsation of hGnRH, the synthesis and release of gonadotropin α subunit and the β subunit of LH is induced. While hGnRH-low frequency pulsatility induces the synthesis and release of the β subunit of FSH (22).

3. Human GnRH receptor signaling in cancer cells

Prostate cancer

Prostate tumors and cell lines derived from these cancers express hGnRH, as well as the hGnRHR; in fact, almost 80% malignant prostate tumors present binding sites for hGnRH (23,24). Assays carried out in human prostate-cancer biopsy, shown that these binding sites were mediated by specific hGnRH receptors (25). Several studies have shown that the hGnRH/hGnRHR system promotes a decrease in cellular proliferation of malignant prostate tumors (26,27). Keeping this in mind, several research groups have been employing hGnRH as antineoplastic drugs for many years (2833).

In tumor cell lines such as LNCaP (androgen-sensitive prostatic-cancer cell line), DU145 cells (a human androgen-independent cancer-cell line) and in PC-82 cells (an androgen-dependent cell line) specific binding sites by hGnRH have been shown (26,27,30). The complete mechanism by which hGnRHR is activated in prostate cancer is unknown; however, the identification of mRNA for hGnRH in LNCaP cells suggests a local paracrine/autocrine system in this tumor cells (34).

Many research groups have investigated the signaling pathway activated by the hGnRH/hGnRHR system to inhibit cell proliferation and ultimate demonstrate cross-talk between growth factor-receptors and hGnRHR. This interaction was observed in LNCaP and DU145 cells, where an association between hGnRHR and epidermal growth factor receptor (EGFR) was shown. In both cell lines the hGnRH stimulation abrogates epidermal growth factor (EGF)-induced transcription factor c-fos expression and reduces the number of EGF-binding sites in membrane. Furthermore, in DU145 cells, inhibition of the EGF receptor phosphorylation and reduction of EGF-binding sites in cell membrane after hGnRH treatment were demonstrated (35). Collectively, these results demonstrated that hGnRHR activation abrogates cell proliferation via EGFR signaling. On the other hand, an important inhibition of the mitogenic action of the insulin-like growth factor 1 (IGF-1) was also observed in DU145 cells after hGnRH-stimulation (36). The inhibitory effects of hGnRH in the proliferation of prostate cancer cells, appears to be mediated via coupling and activating the G protein Gαi, and not by Gαq/11 as observed in gonadotropes (37). In addition to the antiproliferative effect exhibited by the hGnRH/hGnRHR system in prostate cancer it also comprises direct induction of apoptotic signaling (38,39). hGnRH induces apoptosis in DU145 cells involving the activation of c-Jun N-terminal kinase (JNK) by a decrease of protein kinase B (PKB) activity. The activation of JNK could also be mediated by inhibition of the upstream activator of JNK, the mixed-lineage kinase 3 (MLK3) (38). However, details in the mechanisms by which hGnRH induced apoptosis remain to be determined.

Ovarian and endometrial cancer

Temporal and specific expression of hGnRH/hGnRHR has been shown in human ovary cells (40). Radioligand assay carried out in different cell lines and tumor biopsy specimens demonstrated high-affinity binding sites for 125I-labeled hGnRH agonist ([D-Trp6] LHRH) in 70% of primary ovarian cancers as well as in 83% of primary endometrial cancers (41,42). The elucidation of the cellular function of hGnRHR system in extra-pituitary tumor cells has been the goal of many researches. These reports have shown that the expression of both hormone and receptor are able to cause growth inhibition in malignant cells (4245). At the same time, clinical data show that the expression of hGnRH/hGnRHR in epithelial malignant ovary tumors could be considered as a favorable prognostic factor (46).

Although the signaling pathways by which hGnRHR affects cell proliferation in ovarian cancer are still undetermined, it is clear that they are distinct from that in the anterior pituitary (Fig. 1). The specific intracellular signaling cascades that could be coupled to hGnRH in human ovary cancer are the activation of the PKC system. In the human endometrial cancer cell line HHUA, hGnRHR activation was able to downregulate the cellular proliferation via PKC and in human ovarian mucinous cystadenocarcinoma samples, hGnRH agonists also activate PKC protein (47,48). The PKC activation by hGnRHR, could be the link between receptor activation and MAPK kinase cascades to inhibit cell proliferation. In ovarian cancer cells SKOV-3 and OVCAR-3, a pronounced cell proliferation inhibition and activation of extracellular signal-regulated kinase (ERK)1/2 via Gαq/PKC, was demonstrated after hGnRH-stimulation (49).

On the other hand, the antiproliferative action of hGnRHR via ERK1/2 also has been reported as a PKC-independent process suggesting that hGnRHR signaling may vary by cell type (50). In ovarian carcinoma and endometrial carcinoma samples, the activation of the signaling pathways by hGnRHR could be associated with the coupling between the receptor and the Gi protein (51,52). The link between hGnRHR and Gi could explain the differences in the signal transduction pathways activated by hGnRH receptors in malignant tumors and the anterior pituitary. Caov-3 cells shown growth inhibition in response to hGnRH, and also are able to activate several proteins such as adapter protein Src homology 2 domain containing (SHC), superoxide dismutase protein (SOD), MAP kinase kinase protein (MEK) and activation of ERK, via Gi coupling (50).

As mentioned above for prostate cancer, the effects of hGnRHR in mitogenic signaling pathways have also shown cross-talk between this receptor and growth factor receptors in ovary cancer cells. In endometrial (HEC-1A) and ovarian (EFO-21 and EFO-27) cancer cells, hGnRH receptor activation suppresses the phosphorylation of EGFR and inhibits the activation of MAP-kinase/ERK1/2 (53). Similar results were observed in DU-145 cells where hGnRH receptor activation abrogates EGFR-induced c-fos expression and reduces the concentration of EGF-binding sites, resulting in downregulation of cellular proliferation (35). In EFO-21 and EFO-27 cells the inhibition of the EGF receptor is mediated through the coupling between hGnRHR and Gi protein (54). The molecular mechanism for hGnRHR to inhibit the EGFR action may be mediated by direct interaction with intracellular mechanisms activated by EGF as well as by the decrease in the number of EGF receptors present on these cells. For example, in xenografts of OV-1063 cells, hGnRH agonists were able to significantly decrease tumor growth as well as the levels in membrane of EGFR and also its mRNA (55). Furthermore, chronic treatment with hGnRH, in these cells, was able to decrease the levels of insulin-like growth factor I receptor (IGF-1R) (44). A completely new mechanism of GnRH action in endometrial cancer cells has been recently described by Cho-Clark and colleagues (56) demonstrating the participation of a hGnRH metabolite, the GnRH(15), as an active component in the transactivation of EGFR via an orphan GPCR, the GPR101, in Ishikawa cells. Although cross-talk between hGnRHR and growth factor receptors has been clearly demonstrated, it seems to vary in different cellular contexts. Taken together these results show different layers in the complexity of the hGnRH/hGnRHR system regulation in cancer cells. Furthermore, a compound develop by Schally's group, the Dox-14-O-hemiglutarate conjugated to [D-Lys6] GnRH-I (AN-152, AEZS-108; Æterna Zentaris Inc., Quebec, QC, Canada) is currently in phase III clinical trial on ovarian and endometrial cancer due to its promising results in cancer treatment (57).

Breast cancer

Breast cancer is the most common diagnosed cancer and the main global cause of death in women. Approximately, from 75 to 80% of breast cancers are hormone-dependent expressing both estrogen and progesterone receptors (58,59). Approximately 15–20% of breast cancers overexpress the human epidermal growth factor receptor-2 (HER2), and about half of these tumors also express steroid hormone receptors. Unfortunately, 10–15% of breast cancers do not express estrogen or progesterone receptors, nor HER2. These so-called triple-negative breast cancers do not benefit from specific therapies that target these receptors and, therefore, have the worst outcome (59). hGnRH stimulates gonadotropin secretion from the hypothalamus and thereby controls gametogenesis and steroidogenesis in the gonads (60). hGnRH-stimulated gonadotropin secretion can be blocked with antagonists or with a sustained stimulation with agonists, causing the so-called ‘medical castration’ underlying the use of hGnRH analogs to treat hormone-dependent neoplasms (59,60). In addition to expressing the hGnRHR, the tumors also revealed the presence of hGnRH, indicating the existence of an autocrine/paracrine hGnRH/hGnRHR system that might regulate tumor growth and invasiveness (61). Approximately, 50–60% of human breast cancer expresses hGnRHR and it has been highly speculated that activation or inhibition of hGnRHR signaling may directly affect cell growth (6266). Morgan and colleagues (66) demonstrated that hGnRH receptor was expressed in a wide range in 298 primary breast cancers, but most importantly, its expression was significantly higher in patients with triple-negative phenotype.

Initially, it was described that in tumor cells the hGnRHR was exclusively coupled to Gi and consequently inhibit the cAMP accumulation, this action presumably mediates the anti-proliferative effect in these cells (61,67,68). Some reports, however, indicated that for tumor cells the hGnRHR is able to activate several G-proteins in a specific cell background (69). The evidence so far indicates that the anti-proliferative response induced by hGnRHR activation results in apoptosis and G2/M arrest in the cell cycle. This process is mediated via a coordinated dynamic pattern of MAPK, cell cycle, apoptotic and cytoskeletal-related signaling (70).

4. Non-endocrine related cancers

As mentioned above, a broad variety of human cancers, but not normal tissue, express the hGnRH/hGnRHR system, thus, chronic administration of hGnRH agonists are widely and successfully used for the treatment of hormone-dependent tumors. However, study of the hGnRH system in non-hormone-dependent cancers has recently been initiated. For instance, the cutaneous melanoma is still the leading cause of skin cancer deaths in developed countries (71). Moretti and colleagues (72) demonstrated that hGnRHR were expressed in melanoma cells, and the stimulation with a hGnRH agonist had a significant inhibitory effect on tumor progression and neoangiogenesis, by interfering with the activity of growth factors. In multiple myeloma cells RPMI 8226, the use of an hGnRH agonist induces apoptosis and inhibits cell growth by increasing the expression of anti-oncogenes p21 and p53 (73). hGnRH as treatment in colon cancer has been initiated, and preliminary reports suggest an anti-angiogenic property of the hormone (74). As an innovative colon cancer treatment, Schreier and colleagues (75) designed a derivate bioconjugate between a chemotherapeutic agent and a hGnRH analog to target colon cancer cells. Their results demonstrated that the treatment with the bioconjugate changed the protein expression profile of multiple intracellular processes. The use of hGnRH analogs in endometrial cancer had variable responses, some patients respond to therapy but the overall response was suboptimal (76). In some cases, such as in human bladder cancer cells hGnRHR showed a tendency to form nanodomains; however, the effect of any hGnRH analog has not been used in this type of cancer (77).

5. Rescue of misfolded hGnRH receptors

The hGnRHR in its natural form exists as a misfolded protein indicating that the cell intentionally synthesizes conformational misrouted receptors. This characteristic made us hypothesize that the decrease in protein membrane expression is the result of a complex evolutionary reproductive process (78). This hGnRHR that is normally functional become misrouted and then degraded by the cell quality control, therefore ceasing its activity. Among the G-protein coupled receptors, hGnRHR has several specific characteristics including the absence of a terminal carboxy-tail. Another peculiarity of the hGnRHR is the presence of a Lys residue in position 191 of the extracellular loop (EL) 2, in rodents GnRH receptors this orthologous amino acid is absent, making the last protein one residue smaller (327 amino acids). In rodent the lack of this residue confers the GnRH receptor an increased plasma membrane expression (79); therefore, the presence of Lys191 in humans limits the number of receptors exported from the endoplasmic reticulum to the membrane by interfering, primarily, with formation of the Cys14-Cys200 disulphide bridge (Fig. 2) (80). When the disulphide bridge forms, the hGnRHR is recognized by the cell as correctly folded and allows it to be exported to the plasma membrane. As a result, the presence of the Lys191 interferes with the probability of bridge formation and could explain the decrease in trafficking to the plasma membrane. In fact, the deletion of Lys191 from the hGnRHR increases the plasma membrane expression of the protein (81,82). Another structural feature that modulate trafficking of the receptor to the plasma membrane is the formation of a Glu90-Lys121 salt bridge that is essential for the transit through the quality control system of the cell, in fact the Glu90Lys mutant, which led to hypogonadotropic hypogonadism, was retained in the endoplasmic reticulum and represents a loss of function receptor (83,84). Further studies led to the demonstration that this particular mutation was fully rescue by pharmacoperone drugs by the formation of a surrogate bridge from residues Asp98 and Lys121 that can substitute for the original salt bridge that is broken in Glu90Lys mutant (84). Moreover, this human receptor mutation was able to be rescued by four different chemical classes of pharmacoperones that interact identically by creating the same surrogate bridge (85). In fact, pharmacoperones are able to rescue most of the hGnRH receptor mutants, even though mutations appear through the whole receptor sequence, further analysis indicated that these drugs might stabilize the relation between TMD2 and TMD3 domains on the hGnRHR allowing them to pass the quality control of the cell, and thus reaching the membrane. Therefore, it is clear that in human Lys191 is part of a complex motif that results in decreased efficiency in expression (80,82,86).

6. Conclusions

This overview shows that the hGnRH/hGnRHR system activates different signal transduction pathways in gonadotrope and tumor cells, coupling to different G-proteins depending on the cell context. Most interesting is the finding that this hormone/receptor system is present not only in reproductive tissue but also in tumor cells with various degrees of the expression. In some cases the expression of hGnRHR is related to cancer progression; for example, in ovarian carcinomas of early stages the presence of this receptor is higher when compared with advanced stages of carcinoma. Also in prostate cancer loss of hGnRHR expression with tumor progression has been demonstrated (87,88). Moreover, nanomolar concentrations of GnRH II antagonists induce apoptotic cell death in human endometrial, ovarian, and breast cancer in vitro and in vivo, via dose-dependent loss of mitochondrial membrane potential and activation of caspase-3 (89). According to the literature, in several tumor cell lines, the expression level of GnRHR is significantly lower than in pituitary or the gonadotropes (57). The demonstration that hGnRHR exists as a misfolded protein has raised the speculation that in tumor cells this compartmentalization and retention of the receptor in the ER function as a protective mechanism to avoid cell death induced by hGnRHR activation (88,90). In prostate cancer cultures it was demonstrated that IN3 enhances the GnRH agonist apoptotic effect by increasing the hGnRHR in the plasma membrane (88). The use of a careful pulsatile pharmacoperone therapy in a knock-in mouse expressing the hGnRHR mutant E90K is able to restore the mutation from ER retention to the plasma membrane. Also spermatogenetic proteins associated with steroid transport and synthesis, and androgen levels were restored with pharmacoperone administration (91). The hGnRHR plasma membrane increase by pharmacoperones, due to the trafficking of the receptor from the ER, may represent an important research area to evaluate the clinical use of IN3 in tumor cells. Given the clinical utility of hGnRH, further studies of pharmacoperones are necessary to characterize this compound as a step to a more effective and perhaps new therapeutic strategy for cancer patients.

Acknowledgements

The present study was supported by grants FIS/IMSS/PROT/559, FIS/IMSS/PROT/G11/936 (G.M-N) and FIS/IMSS/PROT/G12/1147 (A.A-R).

References

1 

Kakar SS and Jennes L: Expression of gonadotropin-releasing hormone and gonadotropin-releasing hormone receptor mRNAs in various non-reproductive human tissues. Cancer Lett. 98:57–62. 1995. View Article : Google Scholar : PubMed/NCBI

2 

Liu F, Usui I, Evans LG, Austin DA, Mellon PL, Olefsky JM and Webster NJ: Involvement of both G(q/11) and G(s) proteins in gonadotropin-releasing hormone receptor-mediated signaling in L beta T2 cells. J Biol Chem. 277:32099–32108. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Hamm HE: How activated receptors couple to G proteins. Proc Natl Acad Sci USA. 98:4819–4821. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Gilman AG: G proteins: Transducers of receptor-generated signals. Annu Rev Biochem. 56:615–649. 1987. View Article : Google Scholar : PubMed/NCBI

5 

Ford CE, Skiba NP, Bae H, Daaka Y, Reuveny E, Shekter LR, Rosal R, Weng G, Yang CS, Iyengar R, et al: Molecular basis for interactions of G protein betagamma subunits with effectors. Science. 280:1271–1274. 1998. View Article : Google Scholar : PubMed/NCBI

6 

Berridge MJ and Irvine RF: Inositol trisphosphate, a novel second messenger in cellular signal transduction. Nature. 312:315–321. 1984. View Article : Google Scholar : PubMed/NCBI

7 

Limor R, Ayalon D, Capponi AM, Childs GV and Naor Z: Cytosolic free calcium levels in cultured pituitary cells separated by centrifugal elutriation: Effect of gonadotropin-releasing hormone. Endocrinology. 120:497–503. 1987. View Article : Google Scholar : PubMed/NCBI

8 

Tse FW, Tse A, Hille B, Hille B, Horstmann H and Almers W: Local Ca2+ release from internal stores controls exocytosis in pituitary gonadotrophs. Neuron. 18:121–132. 1997. View Article : Google Scholar : PubMed/NCBI

9 

Naor Z, Harris D and Shacham S: Mechanism of GnRH receptor signaling: Combinatorial cross-talk of Ca2+ and protein kinase C. Front Neuroendocrinol. 19:1–19. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Kim Y, Han JM, Park JB, Lee SD, Oh YS, Chung C, Lee TG, Kim JH, Park SK, Yoo JS, et al: Phosphorylation and activation of phospholipase D1 by protein kinase C in vivo: Determination of multiple phosphorylation sites. Biochemistry. 38:10344–10351. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Mitchell R, Robertson DN, Holland PJ, Collins D, Lutz EM and Johnson MS: ADP-ribosylation factor-dependent phospholipase D activation by the M3 muscarinic receptor. J Biol Chem. 278:33818–33830. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Kolch W, Heidecker G, Kochs G, Hummel R, Vahidi H, Mischak H, Finkenzeller G, Marmé D and Rapp UR: Protein kinase C alpha activates RAF-1 by direct phosphorylation. Nature. 364:249–252. 1993. View Article : Google Scholar : PubMed/NCBI

13 

Levi NL, Hanoch T, Benard O, Rozenblat M, Harris D, Reiss N, Naor Z and Seger R: Stimulation of Jun N-terminal kinase (JNK) by gonadotropin-releasing hormone in pituitary alpha T3-1 cell line is mediated by protein kinase C, c-Src, and CDC42. Mol Endocrinol. 12:815–824. 1998.PubMed/NCBI

14 

Mulvaney JM and Roberson MS: Divergent signaling pathways requiring discrete calcium signals mediate concurrent activation of two mitogen-activated protein kinases by gonadotropin-releasing hormone. J Biol Chem. 275:14182–14189. 2000. View Article : Google Scholar : PubMed/NCBI

15 

Seger R and Krebs EG: The MAPK signaling cascade. FASEB J. 9:726–735. 1995.PubMed/NCBI

16 

Maurer RA, Kim KE, Schoderbek WE, Roberson MS and Glenn DJ and Glenn DJ: Regulation of glycoprotein hormone alpha-subunit gene expression. Recent Prog Horm Res. 54:455–484; discussion 485. 1999.PubMed/NCBI

17 

Ben-Menahem D, Shraga-Levine Z, Limor R and Naor Z: Arachidonic acid and lipoxygenase products stimulate gonadotropin alpha-subunit mRNA levels in pituitary alpha T3-1 cell line: Role in gonadotropin releasing hormone action. Biochemistry. 33:12795–12799. 1994. View Article : Google Scholar : PubMed/NCBI

18 

Shraga-Levine Z, Ben-Menahem D and Naor Z: Arachidonic acid and lipoxygenase products stimulate protein kinase C beta mRNA levels in pituitary alpha T3-1 cell line: Role in gonadotropin-releasing hormone action. Biochem J. 316:667–670. 1996. View Article : Google Scholar : PubMed/NCBI

19 

Bliss SP, Navratil AM, Xie J and Roberson MS: GnRH signaling, the gonadotrope and endocrine control of fertility. Front Neuroendocrinol. 31:322–340. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Miller RS, Wolfe A, He L, Radovick S and Wondisford FE: CREB binding protein (CBP) activation is required for luteinizing hormone beta expression and normal fertility in mice. Mol Cell Biol. 32:2349–2358. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Khadra A and Li YX: A model for the pulsatile secretion of gonadotropin-releasing hormone from synchronized hypothalamic neurons. Biophys J. 91:74–83. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Haisenleder DJ, Dalkin AC, Ortolano GA, Marshall JC and Shupnik MA: A pulsatile gonadotropin-releasing hormone stimulus is required to increase transcription of the gonadotropin subunit genes: Evidence for differential regulation of transcription by pulse frequency in vivo. Endocrinology. 128:509–517. 1991. View Article : Google Scholar : PubMed/NCBI

23 

Qayum A, Gullick W, Clayton RC, Sikora K and Waxman J and Waxman J: The effects of gonadotrophin releasing hormone analogues in prostate cancer are mediated through specific tumour receptors. Br J Cancer. 62:96–99. 1990. View Article : Google Scholar : PubMed/NCBI

24 

Halmos G, Arencibia JM, Schally AV, Davis R and Bostwick DG: High incidence of receptors for luteinizing hormone-releasing hormone (LHRH) and LHRH receptor gene expression in human prostate cancers. J Urol. 163:623–629. 2000. View Article : Google Scholar : PubMed/NCBI

25 

Fekete M, Redding TW, Comaru-Schally AM, Pontes JE, Connelly RW, Srkalovic G and Schally AV: Receptors for luteinizing hormone-releasing hormone, somatostatin, prolactin, and epidermal growth factor in rat and human prostate cancers and in benign prostate hyperplasia. Prostate. 14:191–208. 1989. View Article : Google Scholar : PubMed/NCBI

26 

Limonta P, Dondi D, Moretti RM, Maggi R and Motta M: Antiproliferative effects of luteinizing hormone-releasing hormone agonists on the human prostatic cancer cell line LNCaP. J Clin Endocrinol Metab. 75:207–212. 1992.PubMed/NCBI

27 

Dondi D, Limonta P, Moretti RM, Marelli MM, Garattini E and Motta M: Antiproliferative effects of luteinizing hormone-releasing hormone (LHRH) agonists on human androgen-independent prostate cancer cell line DU 145: Evidence for an autocrine-inhibitory LHRH loop. Cancer Res. 54:4091–4095. 1994.PubMed/NCBI

28 

Redding TW and Schally AV: Inhibition of prostate tumor growth in two rat models by chronic administration of D-Trp6 analogue of luteinizing hormone-releasing hormone. Proc Natl Acad Sci USA. 78:6509–6512. 1981. View Article : Google Scholar : PubMed/NCBI

29 

Tolis G, Ackman D, Stellos A, Mehta A, Labrie F, Fazekas AT, Comaru-Schally AM and Schally AV: Tumor growth inhibition in patients with prostatic carcinoma treated with luteinizing hormone-releasing hormone agonists. Proc Natl Acad Sci USA. 79:1658–1662. 1982. View Article : Google Scholar : PubMed/NCBI

30 

Koppán M, Nagy A, Schally AV, Plonowski A, Halmos G, Arencibia JM and Groot K: Targeted cytotoxic analog of luteinizing hormone-releasing hormone AN-207 inhibits the growth of PC-82 human prostate cancer in nude mice. Prostate. 38:151–158. 1999. View Article : Google Scholar : PubMed/NCBI

31 

Arencibia JM, Schally AV, Halmos G, Nagy A, Kiaris H and Klaris H: In vitro targeting of a cytotoxic analog of luteinizing hormone-releasing hormone AN-207 to ES-2 human ovarian cancer cells as demonstrated by microsatellite analyses. Anticancer Drugs. 12:71–78. 2001. View Article : Google Scholar : PubMed/NCBI

32 

Gnanapragasam VJ, Darby S, Khan MM, Lock WG, Robson CN and Leung HY and Leung HY: Evidence that prostate gonadotropin-releasing hormone receptors mediate an anti-tumourigenic response to analogue therapy in hormone refractory prostate cancer. J Pathol. 206:205–213. 2005. View Article : Google Scholar : PubMed/NCBI

33 

Gründker C, Huschmand Nia A and Emons G: Gonadotropin-releasing hormone receptor-targeted gene therapy of gynecologic cancers. Mol Cancer Ther. 4:225–231. 2005.PubMed/NCBI

34 

Limonta P, Dondi D, Moretti RM, Fermo D, Garattini E and Motta M: Expression of luteinizing hormone-releasing hormone mRNA in the human prostatic cancer cell line LNCaP. J Clin Endocrinol Metab. 76:797–800. 1993.PubMed/NCBI

35 

Moretti RM, Marelli MM, Dondi D, Poletti A, Martini L, Motta M and Limonta P: Luteinizing hormone-releasing hormone agonists interfere with the stimulatory actions of epidermal growth factor in human prostatic cancer cell lines, LNCaP and DU 145. J Clin Endocrinol Metab. 81:3930–3937. 1996.PubMed/NCBI

36 

Marelli MM, Moretti RM, Dondi D, Motta M and Limonta P: Luteinizing hormone-releasing hormone agonists interfere with the mitogenic activity of the insulin-like growth factor system in androgen-independent prostate cancer cells. Endocrinology. 140:329–334. 1999.PubMed/NCBI

37 

Maudsley S, Davidson L, Pawson AJ, Chan R, López de Maturana R and Millar RP: Gonadotropin-releasing hormone (GnRH) antagonists promote proapoptotic signaling in peripheral reproductive tumor cells by activating a Galphai-coupling state of the type I GnRH receptor. Cancer Res. 64:7533–7544. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Kraus S, Levy G, Hanoch T, Naor Z and Seger R: Gonadotropin-releasing hormone induces apoptosis of prostate cancer cells: Role of c-Jun NH2-terminal kinase, protein kinase B, and extracellular signal-regulated kinase pathways. Cancer Res. 64:5736–5744. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Kraus S, Naor Z and Seger R: Gonadotropin-releasing hormone in apoptosis of prostate cancer cells. Cancer Lett. 234:109–123. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Choi JH, Gilks CB, Auersperg N and Leung PC: Immuno-localization of gonadotropin-releasing hormone (GnRH)-I, GnRH-II, and type I GnRH receptor during follicular development in the human ovary. J Clin Endocrinol Metab. 91:4562–4570. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Emons G, Ortmann O, Schulz KD and Schally AV: Growth-inhibitory actions of analogues of Luteinizing hormone releasing hormone on tumor cells. Trends Endocrinol Metab. 8:355–362. 1997. View Article : Google Scholar

42 

Völker P, Gründker C, Schmidt O, Schulz KD and Emons G: Expression of receptors for luteinizing hormone-releasing hormone in human ovarian and endometrial cancers: Frequency, autoregulation, and correlation with direct antiproliferative activity of luteinizing hormone-releasing hormone analogues. Am J Obstet Gynecol. 186:171–179. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Emons G, Ortmann O, Becker M, Irmer G, Springer B, Laun R, Hölzel F, Schulz KD and Schally AV: High affinity binding and direct antiproliferative effects of LHRH analogues in human ovarian cancer cell lines. Cancer Res. 53:5439–5446. 1993.PubMed/NCBI

44 

Yano T, Pinski J, Halmos G, Szepeshazi K, Groot K and Schally AV: Inhibition of growth of OV-1063 human epithelial ovarian cancer xenografts in nude mice by treatment with luteinizing hormone-releasing hormone antagonist SB-75. Proc Natl Acad Sci USA. 91:7090–7094. 1994. View Article : Google Scholar : PubMed/NCBI

45 

So WK, Cheng JC, Poon SL and Leung PC: Gonadotropin-releasing hormone and ovarian cancer: A functional and mechanistic overview. FEBS J. 275:5496–5511. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Wilkinson SJ, Kucukmetin A, Cross P, Darby S, Gnanapragasam VJ, Calvert AH, Robson CN and Edmondson RJ: Expression of gonadotrophin releasing hormone receptor I is a favorable prognostic factor in epithelial ovarian cancer. Hum Pathol. 39:1197–1204. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Imai A, Ohno T, Furui T, Takahashi K, Matsuda T and Tamaya T: Gonadotropin-releasing hormone stimulates phospholipase C but not protein phosphorylation/dephosphorylation in plasma membrane from human epithelial ovarian cancer. Int J Gynecol Cancer. 3:311–317. 1993. View Article : Google Scholar : PubMed/NCBI

48 

Shibata S, Sato H, Ota H, Karube A, Takahashi O and Tanaka T: Involvement of annexin V in antiproliferative effects of gonadotropin-releasing hormone agonists on human endometrial cancer cell line. Gynecol Oncol. 66:217–221. 1997. View Article : Google Scholar : PubMed/NCBI

49 

Kim KY, Choi KC, Auersperg N and Leung PC: Mechanism of gonadotropin-releasing hormone (GnRH)-I and -II-induced cell growth inhibition in ovarian cancer cells: Role of the GnRH-I receptor and protein kinase C pathway. Endocr Relat Cancer. 13:211–220. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Kimura A, Ohmichi M, Kurachi H, Ikegami H, Hayakawa J, Tasaka K, Kanda Y, Nishio Y, Jikihara H, Matsuura N, et al: Role of mitogen-activated protein kinase/extracellular signal-regulated kinase cascade in gonadotropin-releasing hormone-induced growth inhibition of a human ovarian cancer cell line. Cancer Res. 59:5133–5142. 1999.PubMed/NCBI

51 

Imai A, Takagi H, Horibe S, Fuseya T and Tamaya T and Tamaya T: Coupling of gonadotropin-releasing hormone receptor to Gi protein in human reproductive tract tumors. J Clin Endocrinol Metab. 81:3249–3253. 1996.PubMed/NCBI

52 

Imai A, Horibe S, Takagi A and Tamaya T: Gi protein activation of gonadotropin-releasing hormone-mediated protein dephosphorylation in human endometrial carcinoma. Am J Obstet Gynecol. 176:371–376. 1997. View Article : Google Scholar : PubMed/NCBI

53 

Emons G, Muller V, Ortmann O, Grossmann G, Trautner U, Stuckrad B, Schulz K and Schally A: Luteinizing hormone-releasing hormone agonist triptorelin antagonizes signal transduction and mitogenic activity of epidermal growth factor in human ovarian and endometrial cancer cell lines. Int J Oncol. 9:1129–1137. 1996.PubMed/NCBI

54 

Gründker C, Völker P and Emons G: Antiproliferative signaling of luteinizing hormone-releasing hormone in human endometrial and ovarian cancer cells through G protein alpha(I)-mediated activation of phosphotyrosine phosphatase. Endocrinology. 142:2369–2380. 2001.PubMed/NCBI

55 

Shirahige Y, Cook C, Pinski J, Halmos G, Nair R and Schally A: Treatment with luteinizing-hormone-releasing hormone antagonist sb-75 decreases levels of epidermal growth-factor receptor and its messenger-RNA in ov-1063 human epithelial ovarian-cancer xenografts in nude-mice. Int J Oncol. 5:1031–1035. 1994.PubMed/NCBI

56 

Cho-Clark M, Larco DO, Semsarzadeh NN, Vasta F, Mani SK and Wu TJ: GnRH-(1–5) transactivates EGFR in Ishikawa human endometrial cells via an orphan G protein-coupled receptor. Mol Endocrinol. 28:80–98. 2014. View Article : Google Scholar

57 

Szabó I, Bősze S, Orbán E, Sipos É, Halmos G, Kovács M and Mező G: Comparative in vitro biological evaluation of daunorubicin containing GnRH-I and GnRH-II conjugates developed for tumor targeting. J Pept Sci. 21:426–435. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Cheang MC, Voduc D, Bajdik C, Leung S, McKinney S, Chia SK, Perou CM and Nielsen TO: Basal-like breast cancer defined by five biomarkers has superior prognostic value than triple-negative phenotype. Clin Cancer Res. 14:1368–1376. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Gründker C, Föst C, Fister S, Nolte N, Günthert AR and 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. 12:R492010. View Article : Google Scholar : PubMed/NCBI

60 

Sedgley KR, Finch AR, Caunt CJ and McArdle CA: Intracellular gonadotropin-releasing hormone receptors in breast cancer and gonadotrope lineage cells. J Endocrinol. 191:625–636. 2006. View Article : Google Scholar : PubMed/NCBI

61 

Limonta P, Montagnani Marelli M, Mai S, Motta M, Martini L and Moretti RM: GnRH receptors in cancer: From cell biology to novel targeted therapeutic strategies. Endocr Rev. 33:784–811. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Fekete M, Wittliff JL and Schally AV: Characteristics and distribution of receptors for [D-TRP6]-luteinizing hormone-releasing hormone, somatostatin, epidermal growth factor, and sex steroids in 500 biopsy samples of human breast cancer. J Clin Lab Anal. 3:137–147. 1989. View Article : Google Scholar

63 

Baumann KH, Kiesel L, Kaufmann M, Bastert G and Runnebaum B: Characterization of binding sites for a GnRH-agonist (buserelin) in human breast cancer biopsies and their distribution in relation to tumor parameters. Breast Cancer Res Treat. 25:37–46. 1993. View Article : Google Scholar : PubMed/NCBI

64 

Moriya T, Suzuki T, Pilichowska M, Ariga N, Kimura N, Ouchi N, Nagura H and Sasano H: Immunohistochemical expression of gonadotropin releasing hormone receptor in human breast carcinoma. Pathol Int. 51:333–337. 2001. View Article : Google Scholar : PubMed/NCBI

65 

Mangia A, Tommasi S, Reshkin SJ, Simone G, Stea B, Schittulli F and Paradiso A: Gonadotropin releasing hormone receptor expression in primary breast cancer: Comparison of immunohistochemical, radioligand and Western blot analyses. Oncol Rep. 9:1127–1132. 2002.PubMed/NCBI

66 

Morgan K, Meyer C, Miller N, Sims AH, Cagnan I, Faratian D, Harrison DJ, Millar RP and Langdon SP: GnRH receptor activation competes at a low level with growth signaling in stably transfected human breast cell lines. BMC Cancer. 11:4762011. View Article : Google Scholar : PubMed/NCBI

67 

Millar RP, Pawson AJ, Morgan K, Rissman EF and Lu ZL: Diversity of actions of GnRHs mediated by ligand-induced selective signaling. Front Neuroendocrinol. 29:17–35. 2008. View Article : Google Scholar

68 

Naor Z and Huhtaniemi I: Interactions of the GnRH receptor with heterotrimeric G proteins. Front Neuroendocrinol. 34:88–94. 2013. View Article : Google Scholar

69 

Sviridonov L, Dobkin-Bekman M, Shterntal B, Przedecki F, Formishell L, Kravchook S, Rahamim-Ben Navi L, Bar-Lev TH, Kazanietz MG, Yao Z, et al: Differential signaling of the GnRH receptor in pituitary gonadotrope cell lines and prostate cancer cell lines. Mol Cell Endocrinol. 369:107–118. 2013. View Article : Google Scholar : PubMed/NCBI

70 

Aguila r-Rojas A, Huer ta-Reyes M, Maya-Núñez G, Arechavaleta-Velásco F, Conn PM, Ulloa-Aguirre A and Valdés J: Gonadotropin-releasing hormone receptor activates GTPase RhoA and inhibits cell invasion in the breast cancer cell line MDA-MB-231. BMC Cancer. 12:5502012. View Article : Google Scholar

71 

Goodson AG and Grossman D: Strategies for early melanoma detection: Approaches to the patient with nevi. J Am Acad Dermatol. 60:719–735; quiz 736–718. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Moretti RM, Mai S, Montagnani Marelli M, Bani MR, Ghilardi C, Giavazzi R, Taylor DM, Martini PG and Limonta P: Dual targeting of tumor and endothelial cells by gonadotropin-releasing hormone agonists to reduce melanoma angiogenesis. Endocrinology. 151:4643–4653. 2010. View Article : Google Scholar : PubMed/NCBI

73 

Wen J, Feng Y, Bjorklund CC, Wang M, Orlowski RZ, Shi ZZ, Liao B, O'Hare J, Zu Y, Schally AV, et al: Luteinizing Hormone-Releasing Hormone (LHRH)-I antagonist cetrorelix inhibits myeloma cell growth in vitro and in vivo. Mol Cancer Ther. 10:148–158. 2011. View Article : Google Scholar

74 

Ghiringhelli F, Isambert N and Ladoire S: Degarelix as a new antiangiogenic agent for metastatic colon cancer? World J Gastroenterol. 19:769–772. 2013. View Article : Google Scholar : PubMed/NCBI

75 

Schreier VN, Pethő L, Orbán E, Marquardt A, Petre BA, Mező G and Manea M: Protein expression profile of HT-29 human colon cancer cells after treatment with a cytotoxic daunorubicin-GnRH-III derivative bioconjugate. PLoS One. 9:e940412014. View Article : Google Scholar : PubMed/NCBI

76 

Carlson MJ, Thiel KW and Leslie KK: Past, present, and future of hormonal therapy in recurrent endometrial cancer. Int J Womens Health. 6:429–435. 2014.PubMed/NCBI

77 

Zhang J, Chtcheglova LA, Zhu R, Hinterdorfer P, Zhang B and Tang J: Nanoscale organization of human GnRH-R on human bladder cancer cells. Anal Chem. 86:2458–2464. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Conn PM, Janovick JA, Brothers SP and Knollman PE: ‘Effective inefficiency’: Cellular control of protein trafficking as a mechanism of post-translational regulation. J Endocrinol. 190:13–16. 2006. View Article : Google Scholar : PubMed/NCBI

79 

Maya-Núñez G, Janovick JA and Conn PM: Combined modification of intracellular and extracellular loci on human gonadotropin-releasing hormone receptor provides a mechanism for enhanced expression. Endocrine. 13:401–407. 2000. View Article : Google Scholar

80 

Conn PM and Janovick JA: Drug development and the cellular quality control system. Trends Pharmacol Sci. 30:228–233. 2009. View Article : Google Scholar : PubMed/NCBI

81 

Janovick JA, Patny A, Mosley R, Goulet MT, Altman MD, Rush TS III, Cornea A and Conn PM: Molecular mechanism of action of pharmacoperone rescue of misrouted GPCR mutants: The GnRH receptor. Mol Endocrinol. 23:157–168. 2009. View Article : Google Scholar :

82 

Maya-Núñez G, Ulloa-Aguirre A, Janovick JA and Conn PM: Pharmacological chaperones correct misfolded GPCRs and rescue function: Protein trafficking as a therapeutic target. Subcell Biochem. 63:263–289. 2012. View Article : Google Scholar : PubMed/NCBI

83 

Maya-Núñez G, Janovick JA, Ulloa-Aguirre A, Söderlund D, Conn PM and Méndez JP: Molecular basis of hypogonadotropic hypogonadism: Restoration of mutant (E(90)K) GnRH receptor function by a deletion at a distant site. J Clin Endocrinol Metab. 87:2144–2149. 2002. View Article : Google Scholar : PubMed/NCBI

84 

Janovick JA, Maya-Nunez G and Conn PM: Rescue of hypogonadotropic hypogonadism-causing and manufactured GnRH receptor mutants by a specific protein-folding template: Misrouted proteins as a novel disease etiology and therapeutic target. J Clin Endocrinol Metab. 87:3255–3262. 2002. View Article : Google Scholar : PubMed/NCBI

85 

Conn PM and Ulloa-Aguirre A: Pharmacological chaperones for misfolded gonadotropin-releasing hormone receptors. Adv Pharmacol. 62:109–141. 2011. View Article : Google Scholar : PubMed/NCBI

86 

Janovick JA, Knollman PE, Brothers SP, Ayala-Yáñez R, Aziz AS and Conn PM: Regulation of G protein-coupled receptor trafficking by inefficient plasma membrane expression: Molecular basis of an evolved strategy. J Biol Chem. 281:8417–8425. 2006. View Article : Google Scholar : PubMed/NCBI

87 

Cheung LW and Wong AS: Gonadotropin-releasing hormone: GnRH receptor signaling in extrapituitary tissues. FEBS J. 275:5479–5495. 2008. View Article : Google Scholar : PubMed/NCBI

88 

Sánchez CA, Mercado AJ, Contreras HR, Cabezas JC, Huidobro CC and Castellón EA: Pharmacoperone IN3 enhances the apoptotic effect of leuprolide in prostate cancer cells by increasing the gonadotropin-releasing hormone receptor in the cell membrane. Anticancer Drugs. 23:959–969. 2012.PubMed/NCBI

89 

Fister S, Günthert AR, Aicher B, Paulini KW, Emons G and 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. 69:6473–6481. 2009. View Article : Google Scholar : PubMed/NCBI

90 

Cheng CK and Leung PC: Molecular biology of gonadotropin-releasing hormone (GnRH)-I, GnRH-II, and their receptors in humans. Endocr Rev. 26:283–306. 2005. View Article : Google Scholar

91 

Janovick JA, Stewart MD, Jacob D, Martin LD, Deng JM, Stewart CA, Wang Y, Cornea A, Chavali L, Lopez S, et al: Restoration of testis function in hypogonadotropic hypogonadal mice harboring a misfolded GnRHR mutant by pharmacoperone drug therapy. Proc Natl Acad Sci USA. 110:21030–21035. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2016
Volume 48 Issue 3

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aguilar-Rojas A, Pérez-Solis MA and Maya-Núñez G: The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review). Int J Oncol 48: 861-868, 2016
APA
Aguilar-Rojas, A., Pérez-Solis, M.A., & Maya-Núñez, G. (2016). The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review). International Journal of Oncology, 48, 861-868. https://doi.org/10.3892/ijo.2016.3346
MLA
Aguilar-Rojas, A., Pérez-Solis, M. A., Maya-Núñez, G."The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review)". International Journal of Oncology 48.3 (2016): 861-868.
Chicago
Aguilar-Rojas, A., Pérez-Solis, M. A., Maya-Núñez, G."The gonadotropin-releasing hormone system: Perspectives from reproduction to cancer (Review)". International Journal of Oncology 48, no. 3 (2016): 861-868. https://doi.org/10.3892/ijo.2016.3346