Looking for a similar answer, essay, or assessment help services?

Simply fill out the order form with your paper’s instructions in a few easy steps. This quick process ensures you’ll be matched with an expert writer who
Can meet your papers' specific grading rubric needs. Find the best write my essay assistance for your assignments- Affordable, plagiarism-free, and on time!

Posted: July 1st, 2024

Gonadotropin Releasing Hormone

Gonadotropin Releasing Hormone

Abstract

Which Citation Styles Can You Handle?

We get a lot of “Can you do MLA or APA?”—and yes, we can! Our writers ace every style—APA, MLA, Turabian, you name it. Tell us your preference, and we’ll format it flawlessly.

 

  1. Introduction:

Gonadotropin-releasing hormone (GnRH) is a decapeptide as shown in Figure 1 produced in the hypothalamus.  It is also known as Luteinizing Hormone-Releasing Hormone (LHRH), gonadoliberin, luliberin, gonadorelin and GnRH I. Despite various functions reported for GnRH in mammals, biochemical, molecular, neuroanatomical, pharmacological and physiological studies have mainly focused on its role as a gonadotrophin-releasing factor (Sealfon, 1997). In human, the reproductive system is regulated by a process that requires the action of hormones in the hypothalamic-pituitary-gonadal (HPG) axis loop. In adult males and females, the hypothalamic pulsatile secretion of the GnRH stimulates the expression and release of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) from the anterior pituitary gland. FSH and LH (gonadotropins) regulate the function of the testes and ovaries (gonads). In both sexes, FSH stimulates gamete production, and LH stimulates the expression of steroid hormones by the gonads. An increased level of steroid hormones inhibits GnRH production through a negative feedback loop (Schally AV, 2000). Over twenty isoforms of GnRH has been identified among vertebrate and protochordate species. Some of these isoforms are shown in Table 1. These structures have been conserved for more than 500 million years of evolution (Karten, 1986b). In most vertebrates at least two, and usually, three, forms of GnRH is present including GnRH-I (GnRH), GnRH-II (also known as chicken GnRH) and GnRH-III (also called lamprey GnRH-III). Studies on different forms of GnRH and its receptor among vertebrates implicated GnRH as a development element in the early evolutionary sequence. In these isoforms of GnRH, the N-terminal (pGlu-His-Trp-Ser) and C-terminal (Pro-Gly-NH2) amino acid sequences of decapeptide are conserved as it is shown in Table 1 (Millar, 2005, Okubo K, 2008). The first identified structure was mammalian GnRH or GnRH-I (Miyamoto K, 1984). The second discovered structure in vertebrate was GnRH-II and isolated from the chicken brain (White SA, 1994). The third isoform has been called GnRH-III and was isolated from fish. As In mammalian, physiological functions and activities are restricted to GnRH I, hence, in the human GnRH-I is referred to as GnRH (Gault PM, 2003), while GnRH-II is also expressed in human(Chen A, 1998). GnRH-II and its structure are uniquely conserved from bony fish to mammals (Millar RP, 2003).

GnRH should specifically bind to the GnRH receptor (GnRH-R), on the surface of gonadotropins cells in the pituitary. This binding results in the stimulation of pituitary gonadotropin release that regulate sperm and ovum maturation and steroidogenesis in the gonads (Harrison et al., 2004). The C and N-termini of the GnRH decapeptide have a role to bind and activate its receptor (Figure 1) (Sealfon, 1997).

GnRH-R is expressed in the pituitary to control the release of gonadotropin hormones LH and FSH (Kakar SS. et al., 1992). GnRH-R is a member of a G-protein coupled receptor (GPCR) family that activates mitogen-activated protein kinase (MAPK) cascades and shows the role of GnRH-R in the regulating of cell growth and proliferation (Zvi Naor, 2000). Hence, expression of GnRH-R has been observed in pituitary adenomas (Alexander JM. and A., 1994) ovary, breast, testis, prostate (Kakar SS. et al., 1992), and granulosa-luteal cells (Derman SG. et al.). GnRH-R is also overexpressed in cancers of gonadal steroid-dependent organs. For instance, in 86% of prostate cancers, 80% of endometrial and ovarian adenocarcinomas, and 50% of breast cancer cases an overexpression of medium to high-affinity binding sites was observed for GnRH and its analogs [Halmos, 2000 #1113;Grundker, 2001 #1119].

Are Writing Services Legal?

Totally! They’re a legit resource for sample papers to guide your work. Use them to learn structure, boost skills, and ace your grades—ethical and within the rules.

GnRH derivatives have been used as therapeutic agents for pharmacological interventions in reproductive dysfunctions including infertility and subfertility, fertility preservation, precious puberty, delayed puberty and endometriosis (Blumenfeld, 2017). The ligands of GnRH-R are extensively used for the treatment of different sex organ-related cancers of women and men, including hormone-sensitive tumors of prostate, breast, ovary, and endometrium. In addition to the primary action of the GnRH receptor ligands in hormone-dependent cancers, a direct growth inhibition has been reported in several hormone-independent cancers of prostate, ovary, and pancreas. It has been shown that this effect of the GnRH analogs was linked to the GnRH receptors overexpression in these tumors (Conn).

This comprehensive overview describes the pivotal roles and mechanisms of GnRH in normal reproduction and GnRH ligands in different complications

  1. GnRH and GnRH-R gene expression in the hypothalamus-pituitary unit

2-1- GnRH gene expression in hypothalamus

What’s the Price for a Paper?

Starts at $10/page for undergrad, up to $21 for pro-level. Deadlines (3 hours to 14 days) and add-ons like VIP support adjust the cost. Discounts kick in at $500+—save more with big orders!

In human, reproductive competence is controlled centrally and linked to the expression of the gene for GnRH (Millar, 2005). A gene on a short arm of chromosome 8 (Region 8p11.2, p21) encodes a 10-kD protein that upon processing yields two biologically peptides of 10 and 56 amino acid residues called GnRH and GnRH-associated peptide (GAP) respectively (Yang-Feng Teresa L. et al., 1985). These peptides are released pulsatile into the portal vascular system that connects the hypothalamus with the pituitary; it has assumed that GAP selectively suppresses releasing of prolactin in the rat, but the physiologic role of GAP has not been well defined. GnRH starts a cascade of neuroendocrine events to change the hormone secretion in pituitary and gonadal steroid output and gametogenesis (RISSMAN et al., 1997).

The gene sequence of the coding region and signal peptide sequence is highly conserved across a wide range of species, differing only in a few amino acids (Clarke and Pompolo, 2005). The 5’ side region of GnRH gene is highly homologous between species (Nelson et al., 1998) and includes two main regions for transcription(Clarke and Pompolo, 2005).

As shown in Figure 2, the primary RNA transcript of GnRH consists of four exons and three introns. The first exon is the 5’ untranslated region (5’-UTR). The second exon encodes the signal peptide, the GnRH decapeptide, the enzymatic amidation and precursor processing site, and the first 11 amino acids of the GnRH-associated peptide (GAP). The third exon encodes the amino acids 12 to 43 of GAP; the fourth exon encodes the remaining amino acids of GAP and the 3’-untranslated region (3’-UTR). After RNA splicing, the most stable and efficient outcome is pro GnRH mRNA which is transferred to the cytoplasm for translation to the peptide (Figure 2). The yield of mature GnRH peptide can be affected by transcription rate, mRNA stability, and post-translational processing (Gore  and Roberts, 1997).

A gene on chromosome 20 encodes GnRH-II which at the amino acid level, shows 70% similarity to  GnRH differing from its structure by three amino acids. This isoform of GnRH is widely identified in the central nervous system, where it seems to act as a neuromodulator of sexual behavior (Chen A, 1998, Millar, 2005, Conn, 1984). GnRH-II is also expressed in different peripheral tissues of the female reproductive system,  like the endometrium, ovary, and placenta as well as in tumors derived from these tissues (Cheon KW, 2001, Millar RP, 2003, Hong IS, 2008).

Is My Privacy Protected?

100%! We encrypt everything—your details stay secret. Papers are custom, original, and yours alone, so no one will ever know you used us.

2-2- GnRH-R gene expression in pituitary

In humans, GnRH receptor (GnRH-R) gene is located on chromosome 4 (4q13.2,3) and includes three exons and two introns which encode for a 328 amino acid protein (Kottler ML, 1995). The type II GnRH-R coding gene consists of a missing nucleotide in the 5’ sequence which possibly caused a frameshift mutation which led to the substitution of a single base change in an Arg codon in exon 2 to a stop codon. So, the type II receptor in humans remains dysfunctional, and its function is now served by GnRH-R for GnRH-II. GnRH-II has shown high affinity to GnRH-R with a different signaling than GnRH (Millar RP, 2003).

Studying of GnRH and its receptor cDNA has been provided that GnRH and GnRH-R are expressed not only in the human reproductive organs but also in liver, heart, skeletal muscle, kidney. This evidence infers that as the main function GnRH and GnRH-R; within the regulation of FSH and LH secretion from the anterior pituitary gonadotropic cells, these may regulate the cellular growth and proliferation while any changes in receptor structure or its expression may lead to uncontrolled growth and proliferation of the cells(Sham S. Kakar and Jennes, 1995).

  1. GnRH endocrine mechanism of action and regulation:

3-1- Endocrine mechanism of action of GnRH and GnRH-R ligands:

Is AI Involved in Writing?

Nope—all human, all the time. Our writers are pros with real degrees, crafting unique papers with expertise AI can’t replicate, checked for originality.

GnRH should specifically bind to its receptor, GnRH-R, to stimulate pituitary gonadotropic cells in the anterior pituitary to release LH and FSH that regulate sperm and ovum maturation and steroidogenesis in the gonads (Conn PM. et al., 1986).

GnRH-R binding and activation are properties of the amino and carboxy-terminal domains of the peptide hormone. The amino-terminal domain is responsible for receptor activation. Other residues outside of the amino-terminal domain can affect receptor activation by altering the peptide conformation (Padula, 2005, Sealfon, 1997).

The GnRH-R is a membrane protein from the family of G-protein-coupled receptors (GPCRs) with seven hydrophobic transmembrane domains (TMD). The amino acid sequence pattern of the GnRH receptor is highly conserved among GPCRs. It contains a single amino acid chain with an extracellular amino-terminal domain and a cytosolic carboxy-terminal domain with no separate terminal signal sequence, and seven hydrophobic parts that are attached to extra and intracellular loops with their α-helical bundle form of membrane spanning (Figure 3). C- terminal of GPCRs family shows the main role to rapid desensitization of receptor, but the lack of a carboxy-terminal tail and an unusual long cytoplasmic loop (Figure 3) in GnRH-R show some unique features in its structure for slow desensitization and internalization (Tsutsumi M, 1992).

Similar to other GPCRs, disulfide bonds between cysteine (Cys) residues are substantial for GnRH-R function. Disruption of the disulfide bonds significantly decreases the affinity of the receptor to GnRH (Figure3) (Fernald and White, 1999). GnRH interacts with the extracellular face of the receptor, inducing a conformational change in the receptor to convert it into its active state. The active conformation spreads signal transduction via associated G-proteins .

Why Are You the Best for Research?

Our writers are degree-holding pros who tackle any topic with skill. We ensure quality with top tools and offer revisions—perfect papers, even under pressure.

Activation of GnRH-R by GnRH causes indirect stimulation of phospholipase C (PLC), which hydrolyzes membrane phosphoinositides to inositol1,4,5-trisphosphate (IP3). IP3 mobilizes intracellular Ca2+ (Colonna, 2008). GnRH increases Ca2+ entry via voltage-operated channels into gonadotropic cells. This intensification in the concentration of cytosolic Ca2+ is primarily responsible for the LH release (Hislop JN et al., 2000).

The results of binding GnRH to its receptor is followed by two cellular responses. First, the rapid influx of Ca2+ into cells the and activation of calmodulin (an intermediate calcium-binding messenger protein). Second, stimulation of the cell production of specific membrane-associated lipid-like diacylglycerols as a second messenger to activate the enzyme protein kinase C (PKC). PKC activates the MAPK cascades which increase LH and FSH transcription in pituitary cells(Harrison et al., 2004). Consequently, activated calmodulin and activated PKC enhance the release of gonadotropins. Simultaneous, diacylglycerols amplify the action of calmodulin to increase the release of gonadotropins(Conn, 1986).

In pituitary gonadotropic cells, GnRH-R activates some other pathways like cytoskeletal remodeling and cyclic AMP-dependent protein kinase A (cAMP/PKA) pathways signaling (Perrett and McArdle, 2013, Bhave and Hoffman, 2003). cAMP/PKA pathway possibly activate gonadotropin subunit transcription that has cAMP response elements (CREs) as a promoter subunits (25). Cytoskeletal remodeling amplifies the tyrosine phosphorylation status of a series of cytoskeleton-associated proteins that have an anti-apoptotic effect on neurons and prevent neurodegeneration (Perrett and McArdle, 2013, Hsueh and Jones, 1983).

  1.          Regulation of gonadotropins and GnRH:

It is a poorly understood how GnRH secretion in the hypothalamus and GnRH-R expression in the pituitary neurons are regulated. GnRH secretion can be influenced by a range of factors, such as stress, nutrition and gonadal feedback (Clarke and Pompolo, 2005).

Who Writes My Assignments?

Experts with degrees—many rocking Master’s or higher—who’ve crushed our rigorous tests in their fields and academic writing. They’re student-savvy pros, ready to nail your essay with precision, blending teamwork with you to match your vision perfectly. Whether it’s a tricky topic or a tight deadline, they’ve got the skills to make it shine.

  1.      Regulation of gonadotropins secretion at the pituitary level

GnRH is released in a pulsatile manner from the hypothalamus and binds to the GnRH-R on the cell surface of the gonadotropic cells. The secretion patterns of FSH and LH is dependent on the differential GnRH pulse frequencies and amplitudes (Savoy-Moore RT, 1987). It is not clearly known how the gonadotropic cells decode the pulsatile GnRH signal. However, it is known that multiple distinct signaling pathways are activated by varying frequencies of pulsatile GnRH. Both stimulatory and repressive transcription factors are activated by pulsatile GnRH. Increasing frequencies results in preferential secretion of LH while decreasing frequencies leads to FSH release. Apart from the fundamental role of slow and fast GnRH frequencies in activating signaling pathways for FSH and LH synthesis, other pathways such as inhibins and activins, sex steroid feedback, or epigenetic regulation are involved in the process (Thompson and Kaiser, 2014).

Intermediate-to-high GnRH pulse frequencies favor the transcription of the ß-subunit of LH, while the lower GnRH pulse frequencies result in the greater production of FSH ß-subunit. In children, GnRH secretion occurs at a low amplitude and frequency and both amplitude and frequency increase during pubertal maturation. In men, secretion of GnRH is varied with pulses occurring approximately every two hours keeping a relatively consistent pattern throughout adult life. In women, the pattern of GnRH pulses changes during the menstrual cycle to maintain cyclic ovulation. The pulse frequencies increase during the follicular phase and before ovulation, which contributes to the generation of the preovulatory gonadotropin surge (Ferris H.A., 2006). Pulse frequencies are reduced post-ovulation phase due to feedback by estradiol and progesterone from the corpus luteum (Marshall JC, 1993).

GnRH-R regulation is also depended on GnRH pulse frequency secretion. At high GnRH pulse frequencies, a Regulator of G-protein signaling (RGS)-2 inhibits signaling of the GnRH-R via a negative transcription-dependent feedback on upstream inputs (Perrett and McArdle, 2013). While GnRH interacts with GnRH-R to induce gonadotropin release, it regulates the expression of GnRH-R. The pituitary cells response to GnRH pulses to change numbers receptor due to different states of the ovulation cycle (Clarke and Pompolo, 2005). The regulation of the number of GnRH-R is also affected by gonadal steroids, inhibin, gonadotropins, and GnRH (Kovacs and Schally, 2001).  Dysfunctional GnRH pulsatility has seen in polycystic ovary syndrome, obesity, hypothalamic amenorrhea and pubertal disorders (Marques P, 2000).

The energy stress due to periods of starvation/energy-deprivation suppresses GnRH pulsatility (De Souza MJ, 2007). This suppression reduces pituitary secretion of the gonadotropins and leading to hypothalamic hypogonadism (Compagnucci C and Cebral E, 2002, Huang W, 2008 ). GnRH neurons can directly sense nutritional signals like insulin and ghrelin level (Farkas I, 2013, Evans MC, 2014, Evans and Anderson, 2017).

Will My Paper Be Unique?

Guaranteed—100%! We write every piece from scratch—no AI, no copying—just fresh, well-researched work with proper citations, crafted by real experts. You can grab a plagiarism report to see it’s 95%+ original, giving you total peace of mind it’s one-of-a-kind and ready to impress.

  1.   Regulation of GnRH by Gonadal Feedback

Estrogen and progesterone level in females and testosterone in males have been shown to play a major role in the GnRH regulation. In males, testosterone has been shown to have a strong and direct negative feedback on the GnRH secretion from the hypothalamus. Under certain circumstances, the steroids directly affect the GnRH release at the pituitary gland. Furthermore, FSH production is inhibited by the hormone inhibin, which is released by the testes (Figure 4) (Tilbrook AJ1, 1995).

In females, at the beginning of the menstrual cycle known as follicular phase, GnRH is secreted pulsatile from the mediobasal of the hypothalamus and internalized to gonadotropic cells in the anterior pituitary via its receptors. Then GnRH stimulates pulsatile releasing of FSH and LH which are leading to the follicular growth and the ovarian secretion of estrogen. The high levels of estrogen and the LH surge induce ovulation (Alyasin A. et al., 2016). After releasing of the ovum, at the beginning of the luteal phase, the secretion of progesterone increases while estrogen level drops down. At the middle of luteal phase increase at estrogen level and progesterone surges down regulates GnRH. At the end of the luteal phase, a decrease in the plasma progesterone levels leads to an increase in GnRH pulse frequencies and subsequent LH release (Clarke, 1995, Moenter, 1991, Clarke, 1987).

In males, GnRH stimulatory actions on the pituitary induce releasing of FSH and LH; FSH stimulates the production of sperm cells by signaling to undergo meiosis while LH stimulates the Leydig cells and the testes to synthesize and secrete testosterone to encourage sperm production and secondary sexual characteristics (Tilbrook and Clarke, 1995).

The effects of sex steroids on the GnRH regulation change the patterns of LH secretion in males and females during the gametogenesis cycles. It had been hypothesized that regulation of GnRH, LH, and sex steroid hormones are a direct cycle, i.e. GnRH pulses regulate LH pattern, LH pattern regulates sex steroid hormones release, and sex steroid hormones regulate GnRH (Clarke and Pompolo, 2005). However, studies showed that GnRH neurons neither express progesterone or androgen receptor (Skinner, 2001) nor do they express estrogen receptor (ER) (Herbison, 2001). Hence, the effects of steroid hormones on GnRH synthesis supposed to involve neuronal intermediaries as represented in Figure 4 (Tilbrook, 2002. ).

Can You Use Any Citation Format?

Yep—APA, Chicago, Harvard, MLA, Turabian, you name it! Our writers customize every detail to fit your assignment’s needs, ensuring it meets academic standards down to the last footnote or bibliography entry. They’re pros at making your paper look sharp and compliant, no matter the style guide.

Estradiol (E2) regulates GnRH secretion through positive feedback on GnRH neurons via direct and indirect pathways. E2 exerts feedback directly on GnRH neurons through rapid membrane-associated responses that involve G-proteins STX-R and GPR30 shown in Figure2. It has been demonstrated that membrane-associated receptors including GPR30 and a diphenyl acrylamide compound (STX)-sensitive receptors have an intermediary role for the prompt direct action of estradiol in the GnRH-secreting hypothalamic neurons (Figure 2)(Kenealy and Terasawa, 2011). E2 signal to GnRH neurons indirectly through various neural afferent networks and neuromodulators shown in Figure3.

  1.   Regulation of GnRH secretion by neuronal intermediaries

Neurotransmitter systems and ionotropic GABA-ergic neurons have been shown to moderate the effects of estrogen (Nunemaker, 2002). This indicates while gonadal steroids such as estrogen and progesterone are the regulators of GnRH secretion, they might not have direct effects on GnRH-secreting neurons. Hence, the effects of steroid hormones on the GnRH synthesis is believed to involve neuronal intermediaries. Kisspeptin plays a key role as GnRH regulator. It is released in the infundibular nucleus and acts as an intermediate to induce negative feedback in estrogen release in humans (Tilbrook, 2002. ). Kisspeptin is a strong stimulator of the HPG axis in both animal models and humans. Kisspeptin is known as a hypothalamic peptide encoded by the KISS1 gene, and acts as a neuromodulator upstream of GnRH and sensitizes to sex steroid feedback and metabolic strings. Expression of the KISS1 gene is under the control of both estrogens and androgens (Rometo AM, 2007). Kisspeptin is now well known to be essential for the control of fertility and regulation of the sexual maturity via sex hormone-mediated secretion of gonadotropins (Pinilla L, 2012).

Kisspeptin directly sends signals to GnRH neurons, which express KISS1R. The location of kisspeptin neurons within the hypothalamus is species-specific. Kisspeptin neurons in humans are within the preoptic area (POA) and the infundibular nucleus. Kisspeptin neurons in the infundibular nucleus co-express neurokinin B and dynorphin (KNDy neurons), which auto-synoptically regulate kisspeptin secretion (through the neurokinin B receptor and kappa opioid peptide receptor). In humans, infundibular KNDy neurons transmit negative and positive feedbacks. The role of POA kisspeptin neurons in sex steroid feedback is not clear yet (Skorupskaite et al., 2014).

Regarding Kisspeptin and KNDy neurons key role in the control of GnRH release, a therapeutic approach to modify the activity of the GnRH pulse frequency based on the utilization of kisspeptin, Neurokinin B or dynorphin analogs has emerged. This approach could lead to an inefficient GnRH pulse modulation which is more efficient than a complete activation/ suppression by analogs, thus reducing the possible GnRH analogue-related side effects (Skorupskaite et al., 2014).

Can I Change My Order Details?

For sure—you’re not locked in! Chat with your writer anytime through our handy system to update instructions, tweak the focus, or toss in new specifics, and they’ll adjust on the fly, even if they’re mid-draft. It’s all about keeping your paper exactly how you want it, hassle-free.

  1.   Other regulating factors

The role of other hormones in the GnRH regulation has also been studied. Serotoninergic system is shown to have a negative feedback on the GnRH biosynthesis, but still, the effect of serotonin on GnRH mRNA expression in females is unknown (Li, 1992). Additionally, noradrenaline represents dual effects on GnRH, and LH secretion according to steroid level status (Kalra, 1993).

The neuroactive substances that are well studied in the regulation of GnRH gene expression are glutamate, catecholamine norepinephrine, and γ-aminobutyric acid (GABA). Early studies showed that the inhibitory neurotransmitter GABA is an inhibitor of GnRH secretion (Herbison, 1998), but later studies demonstrated a stimulatory role for the GABA in  GnRH secretion through activating Ca2+ channels and increasing level of Ca2+ (Figure 2).  The effect of GABA on GnRH mRNA levels is not clear (Clarke and Pompolo, 2005). The excitatory neurotransmitter glutamate involves in the regulation of GnRH secretion. Glutamate stimulates GnRH release from hypothalamic neurons by activating the release of nitric oxide (NO).  NO increases the level of cGMP by activating guanylate cyclase which leads to stimulating GnRH secretion (Figure 2) (Dhandapani, 2000).

It has been reported that PKA activators (Wetsel WC, 1993), protein kinase C (PKC)(Eraly SA, 1995), Ca2+ (Krsmanovic LZ, 1992), or PLC bypass the binding of the ligand to its receptor and directly activate the intracellular second messenger cascades. These intermediates are found to have stimulatory effects on the GnRH secretion (Gore  and Roberts, 1997).

 

How Do I Order a Paper?

It’s a breeze—submit your order online with a few clicks, then track progress with drafts as your writer brings it to life. Once it’s ready, download it from your account, review it, and release payment only when you’re totally satisfied—easy, affordable help whenever you need it. Plus, you can reach out to support 24/7 if you’ve got questions along the way!

 

  1. Clinical application of GnRH and its analogs

GnRH analogs have been used as therapeutic agents for pharmacological interventions in reproductive dysfunctions including infertility and subfertility, fertility preservation, precious puberty, delayed puberty and endometriosis. GnRH analogs are also widely prescribed for reduction of gonadotoxicity side effects of chemotherapy in young women, assisted reproduction (ART)/in vitro fertilization (IVF), benign prostate hyperplasia, uterine leiomyomata, polycystic ovary syndrome (PCOS), and precocious puberty (Blumenfeld, 2017). The ligands of GnRH-R are extensively used for the treatment of different sex organ-related cancers of women and men, including hormone-sensitive tumors of prostate, breast, ovary, and endometrium (Conn).

Clinical use of GnRH is summarized in Table 2.

    5-1- GnRH analogs – Endocrine activity

How Quick Can You Write?

Need it fast? We can whip up a top-quality paper in 24 hours—fully researched and polished, no corners cut. Just pick your deadline when you order, and we’ll hustle to make it happen, even for those nail-biting, last-minute turnarounds you didn’t see coming.

The desired aim of the treatment with GnRH analogs is to suppress the gonadotropin surge and sex steroid secretion by GnRH-R desensitization (Kovacs and Schally, 2001). Overstimulation of GnRH-R by GnRH agonists and blocking the receptor to inhibit gonadotropin release by GnRH antagonists down-regulate GnRH-R, which results in a significant reduction in endogenous sex hormones.

The molecular structure of GnRH is simple enough to synthesize thousands of active chemical analogs (agonistic and antagonistic) (Conn PM. et al., 1986, Karten, 1986a). Advanced technology in solid-phase peptide synthesis provides the automated production of short peptides (Merrifield, 1966). It has been estimated that over thousand different analogs of GnRH have subsequently been synthesized (Karten, 1986b).

The C-terminal of the GnRH decapeptide is essential for the binding to the receptor while its N-terminal is critical for both receptor binding and activation. Substitution of the primary amino acids of GnRH initially targeted the C-terminus because the N-terminal domain residues are mostly responsible for receptor activation (His and Trp) as shown in Figure 1 and Table 3. Modification of residues outside of the N-terminal domain can affect receptor activation (Padula, 2005, Sealfon, 1997). Studies demonstrated that amino acids in positions 1 and 4–10 are also essential to provide the proper conformation (Schally, 1999).

Rapid cleavage of the Gly-Leu bonds in GnRH has been shown to result in a short half-life of 2–4 min. Thus, to increase the stability of the GnRH analogs, the Gly at the 6th  position of decapeptide has been substituted. This leads to a higher plasma half-life compared to the native peptide (Maggi et al., 2016).

5-1- 1- GnRH agonist

Can You Handle Tough Topics?

Absolutely—bring it on! Our writers, many with advanced degrees like Master’s or PhDs, thrive on challenges and dive deep into any subject, from obscure history to cutting-edge science. They’ll craft a standout paper with thorough research and clear writing, tailored to wow your professor.

Agonists of GnRH are pharmacologically more active than GnRH, and they are usually modified at the 6th and 10th position of the GnRH peptide chain to achieve an extended half life and receptor binding affinity (Vickery, 1986). Substitution of Gly at the 6th position by D-amino acid increases receptor binding affinity by stabilizing the ß-turn conformation that has been shown to be essential for ligand interaction (Laimou, 2010). To increase the agonist binding affinity to GnRH-R, Gly at the 10th position has been replaced by ethylamide residue that binds to Pro9. This substitution enhances the potency of the peptide for inducing receptor because Pro-alkylamine moiety induces a prolonged duration of action (Karten, 1986b). GnRH agonists such as triptorelin, leuprolide (Wilson AC, 2007), buserelin, and goserelin, have been used worldwide for nearly two decades (Laimou et al., 2012). The sequences of these GnRH agonists are shown in Table 4.

Like GnRH, GnRH agonists stimulate gonadotropins biosynthesis and secretion which causes an increase in the number of GnRH-R, gonadotropin and sex steroids secretion (Perrett and McArdle, 2013). This initial sex steroid surge is known as “flare-up reaction” and results in the down regulation of GnRH release (as shown in Figure) and followed by desensitization of the receptor in gonadotrophs (J. Waxman, 1985). While an acute injection of the GnRH agonists increases the number of GnRH-R and induces the release of LH and FSH, chronic administration results in inhibitory effects. Chronic administration of GnRH agonists continuously stimulates the receptors in the pituitary gonadotropes which then downregulates and desensitizes receptors in gonadotrophs followed by LH surge and sex steroid levels inhibition (Kovacs and Schally, 2001, Wilson AC, 2007).

In prostate cancer, the stimulatory actions of the agonists on the pituitary induces an initial release of LH and FSH, which acts to increase plasma testosterone levels until downregulation has occurred and eventual castrate levels of testosterone is reached. The flare-up response temporary worsens the existing signs with increased bone pain in up to 20% of patients. Pretreatment with an anti-androgens can reduce the flare-up side effect of GnRH agonist (Padula, 2005).

As GnRH agonist showed well controlled ovarian stimulation (COS), triptorelin is administrated for the treatment of infertility related to PCOs(Chang RJ, 1984).

In breast cancer therapy the lowest level of estrogen by ovarian steroidogenesis suppression within chemotherapy is desired. In premenopausal women, a combination therapy of GnRH agonists goserelin (Zoladex) and chemotherapy drugs like tamoxifen have shown significant therapeutic effects via causing estrogen deprivation (A.V. Schally, 1976). In advanced breast cancer, GnRH agonist within temporary convert the premenopausal women to postmenopausal can increase the chemotherapy drug choices and survival (Robertson and Blamey, 2003).

How Do You Match Professor Expectations?

We follow your rubric to a T—structure, evidence, tone. Editors refine it, ensuring it’s polished and ready to impress your prof.

The primary route of administration of GnRH agonist is intravenous (IV), intramuscular (IM), and subcutaneous (SC). While the oral route is the preferred route of administration by patients, poor membrane permeability and susceptible to gastrointestinal peptidase degradation, making the oral administration of GnRH agonists unsuitable with less than 0.1% bioavailability (Iqbal et al., 2012). Developing oral administration route of GnRH agonist is under further investigation (Moradi et al., 2015). The intranasal route is well tolerated in humans. However, intranasal administration is inefficient and variable with necessitating frequent large doses (Padula, 2005).

5-1-2- GnRH antagonist

GnRH antagonists produce a competitive and permanent blockade of GnRH-R and immediately stop the gonadotropins and sex steroids release. Hence, compared with the agonists their onset of action is significantly faster(Kovacs and Schally, 2001, Karten, 1986b, Hahn, 1981). GnRH antagonists have a long half life and high affinity to bind to the GnRH receptors but without activating them (Karten, 1986b). Studies showed that any changes at positions 1 and 10 notably increase the receptor binding efficiency of the peptide and substitutions at positions 2 and 3 inhibit gonadotropin release activity (A.V. Schally, 1976). It was also considered that the insertion of D-Arg or other related basic amino acid residues at 6th  position of GnRH antagonists intensify its potential (J.E. Rivier, 1986). However, this modification induced histamine liberation resulting in transient edema and other anaphylactoid reactions. Substitution of the neutral residues like D-Cit at 6th  position in cetrorelix has not shown any anaphylactoid reactions and more confidence for clinical use. Cetrorelix also shows the higher suppression effect rate than other GnRH antagonists (Schally, 1999).

GnRH antagonists such as Ganirelix, Cetrorelix, and Abarelix has been introduced into the clinical practice (Laimou et al., 2012). The amino acid sequence of some GnRH antagonists is shown in Table 5.

How Do You Edit My Work?

Send us your draft and goals—our editors enhance clarity, fix errors, and keep your style. You’ll get a pro-level paper fast.

However, studies demonstrated that chronic administration of cetrorelix also induces desensitization of gonadotropes, down-regulation of GnRH-R in the pituitary, and reduce the levels of mRNA for GnRH-R. Thus, the down-regulation of GnRH-R in pituitary by antagonists of  GnRH seems to be similar to those resulting from the agonists without flare-up reaction. Due to the absence of flare-up response within rapid down-regulation, there is no need for any combinatorial treatment with anti-sex steroids in GnRH antagonist treatments. GnRH antagonist is suggested to be more suitable for clinical practice than agonists (Al-Inany et al., 2011).

In prostate cancer, antagonists induce a down-regulation of GnRH-R in pituitary during long term administration of cetrorelix in men (Pinski J, 1996, Behre HM, 1997). In patients with advanced prostate cancer treatment with cetrorelix decreased their testosterone and prostate specific antigen (PSA) level and prostate size as measured by ultrasonography (Gonzalez–Barcena D, 1994).

Administration of GnRH antagonist can rapidly suppress LH and testosterone levels and inhibit ovarian hyperstimulation in women with polycystic ovarian disease (Hayes FJ, 1998).

  1. Significant in preclinical investigation of GnRH analogs

The overexpression of GnRH-R has been well reported in many cancer tumors especially breast, ovary, endometrial and prostate cancer. Many studies have targeted GnRH or conjugation of the GnRH analog for anti-cancer therapy due to the high expression levels of GnRH receptors in some various malignant human tumor cell lines. Direct anticancer activity of GnRH analogs has been reported (Montagnani Marelli, 2007). Activation of GnRH-R in these cell lines has shown strong antiproliferative, antimetastatic, and antiangiogenic activity (Parborell F, 2008). GnRH or its analogs can be conjugated as targeting ligands to drugs and nanocarriers containing cytotoxic drugs agents. The combination can directly target cancer cells with overexpressed GnRH-R, by increasing toxic effects in tumor and spare benign cells from undesired damage (Schally AV, 2004). The uptake of the cytotoxic drug or nanocarrier occurs via an interaction between GnRH and its receptor and then endocytosis by the cell membrane. As normal cells with no overexpression of GnRH-R could not uptake the drug or nanocarrier, the side effects of the cytotoxic drug are significantly reduced (Arangoa MA, 2001).

AEZS-108 (formerly known as AN-152) is a GnRH agonist conjugation with doxorubicin that has shown anti-cancer activity in phase II clinical trial of gynecological cancers and a promising result for prostate cancer (Engel J, 2012).

Can You Brainstorm Topics?

Yep! We’ll suggest ideas tailored to your field—engaging and manageable. Pick one, and we’ll build it into a killer paper.

EP-100 is another successful GnRH agonist conjugated compound for a solid tumor in phase II clinical trial. In this formulation GnRH linked to a cationic membrane-disrupting peptide that interacts with the negatively charged membrane of cancer cells and inducing apoptosis through the plasma and mitochondrial membrane lysis (Curtis et al., 2014).

TB1 is a triptorelin (T) as GnRH agonist conjugated to antimicrobial peptides (B1) with anti cancer activity and has been investigated in vitro. TB1 showed anti cancer effects on breast, prostate, cervical, hepatic, leukemia, gastric epithelial and kidney cancer cell lines which had been proven to overexpress GnRH-H on their membrane (Deng et al., 2015).

GnRH has been conjugated for targeted delivery of encapsulated cisplatin in dextran nanoparticle to breast cancer to suppress cancer growth and metastasis in mice model. GnRH-nanoparticles has shown great potential for the targeted chemotherapy of metastatic breast cancer. (Li M, 2015).

The conjugation of the GnRH analog to lipophilic and glycosyl moieties has demonstrated to increase the stability and membrane permeability of the peptide, which induced variable antiproliferative effects in hormone dependent prostate and ovarian cancer cell proliferation (Varamini et al., 2017, Goodwin et al., 2015).

Due to the poor stability and low oral bioavailability of GnRH, no oral analog for clinical use exist. Lactose conjugated of GnRH not only has resulted in increased stability of peptide but also enhanced the oral bioavailability in rats significantly (Moradi et al., 2015).

A novel approach of therapeutical application of GnRH is to use vaccination against GnRH in which anti-GnRH antibodies neutralize GnRH and suppress LH and FSH secretion and decrease sex steroids levels. Vaccination with anti-GnRH has suggested the treatment for hormone-dependent cancer and as a potential alternative to surgical castration (Chang et al., 2015). Some fusion proteins as a toxoid carrier have been developed for the conjugation to anti-GnRH antibodies (Miller et al., 2008). A recombinant fusion protein consisting of GnRH and diphtheria toxoid was successful in phase I/II clinical trials of prostate cancer (Parkinson et al., 2004). A novel design of an anti-GnRH vaccine with assembling of aCD4 T-cell epitope (an influenzaHA2 light chain), the self-peptide GnRH (the B-cell epitope), and lipid moiety to improve the immunogenicity of the anti-GnRH vaccine constructs, has shown significant results for gynecological cancer in vivo and prostate cancer in vitro (Chang et al., 2015).

The further preclinical investigation is summarized in table 5.

Do You Offer Fast Edits?

Yes! Need a quick fix? Our editors can polish your paper in hours—perfect for tight deadlines and top grades.

  1. Conclusion:

Hypothalamic pulsatile secretion of the GnRH is a key control of the reproductive system by stimulating the expression and release of FSH and LH from the anterior pituitary gland to regulate the function of the gonads for gametogenesis and steroidogenesis. As a key control, GnRH has a major therapeutic role in any reproductive dysfunctions and sex-hormone dependent disease. The main therapeutic aim of GnRH is to suppress the gonadotropin surge and sex steroid secretion. Since the native GnRH is not stable, synthesize the analog of GnRH well studied for clinical use. Temporary Overstimulation of GnRH-R by GnRH agonists and permanently blocking the receptor to inhibit gonadotropin release by GnRH antagonists down-regulate GnRH-R, which results in a significant reduction in endogenous sex hormones. Targeted drug delivery investigations based on GnRH and its receptor for hormone dependent these cancers have emerged since overexpression of GnRH-R well reported in these cancer tumors. Vaccination against GnRH is a novel approach of therapeutical application of GnRH under further investigations.

A.V. SCHALLY, A. J. K., D.H. COY, EDWARD T. 1976. LH-releasing hormone and its analogues: recent basic and clinical investigations. International journal of fertility, 21, 1-30.

AL-INANY, H. G., YOUSSEF, M. A. F. M., ABOULGHAR, M., BROEKMANS, F., STERRENBURG, M., SMIT, J. & ABOU-SETTA, A. M. 2011. GnRH antagonists are safer than agonists: an update of a Cochrane review. Human Reproduction Update, 17, 435-435.

ALEXANDER JM. & A., K. 1994. Gonadotropinreleasing hormone receptor mRNA expression by human pituitary tumors in vitro. J Clin Invest 93.

ALYASIN A., MEHDINEJADIANI S. & GHASEMI M. 2016. GnRH agonist trigger versus hCG trigger in GnRH antagonist in IVF/ICSI cycles: A review article. Int J Reprod BioMed, 14, 557-566.

Can You Start With an Outline?

Sure! We’ll sketch an outline for your approval first, ensuring the paper’s direction is spot-on before we write.

ARANGOA MA, C. M., RENEDO MJ, PONCHEL G, IRACHE JM. 2001. Gliadin nanoparticles as carriers for the oral administration of lipophilic drugs. Relationships between bioadhesion and pharmacokinetics. . Pharmaceutical Research, 18, 1521-1527.

BEHRE HM, K. S., PU¨HSE G, REISSMANN T, NIESCHLAG E 1997. High loading and low maintenance doses of a gonadotropin-releasing hormone antagonist effectively suppress serum luteinizing hormone, follicle-stimulating hormone, and testosterone in normal men. J Clin Endocrinol Metab, 82, 1403-1408.

BHAVE, S. V. & HOFFMAN, P. L. 2003. Phosphatidylinositol 3′-OH kinase and protein kinase A pathways mediate the anti-apoptotic effect of pituitary adenylyl cyclase-activating polypeptide in cultured cerebellar granule neurons: modulation by ethanol. Journal of Neurochemistry, 88, 359-369.

BLUMENFELD, Z. 2017. Investigational and experimental GnRH analogs and associated neurotransmitters. Expert Opin Investig Drugs, 26, 661-667.

CHANG, C., VARAMINI, P., GIDDAM, A. K., MANSFELD, F. M., D’OCCHIO, M. J. & TOTH, I. 2015. Investigation of Structure–Activity Relationships of Synthetic Anti‐Gonadotropin Releasing Hormone Vaccine Candidates. ChemMedChem, 10, 901-910.

CHANG RJ, L. L., MELDRUM DR, DE FASIO J, LU JKH, VALE WW, RIVIER JW, JUDD HL. 1984. Steroid secretion in polycystic ovarian disease after ovarian suppression by a long-acting gonadotropin-releasing hormone agonist. J Clin Endocrinol Metab, 56, 897-903.

CHEN A, Y. D., BEN-AROYAN, KAGANOVSKY E,OKON E, KOCH Y.A 1998. second isoform of gonadotropin-releasing hormone is present in the brain of human and rodents. . FEBS Lett 435, 199– 203.

CHEON KW, L. H., PARHAR IS, KANG IS. 2001. Expression of the second isoform of gonadotrophin-releasing hormone (GnRH-II) in human endometrium throughout the menstrual cycle. . Mol Hum Reprod 7, 447– 452.

Can You Add Charts or Stats?

Definitely! Our writers can include data analysis or visuals—charts, graphs—making your paper sharp and evidence-rich.

CLARKE, I. J. 1987. New concepts in gonadotrophin-releasing hormone action on the pituitary gland. . Semin.Reprod. Endocrinol., 5, 345–352.

CLARKE, I. J. 1995. Evidence that the switch from negative to positive feedback at the level of the pituitary gland is an important timing event for the onset of the preovulatory surge in LH in the ewe. J. Endocrinol. , 145, 271–282.

CLARKE, I. J. & POMPOLO, S. 2005. Synthesis and secretion of GnRH. Anim Reprod Sci, 88, 29-55.

COLONNA, M. 2008. Below the NK-cell surface: PIP2. Blood, 111, 3916-3916.

COMPAGNUCCI C, C. G., LOMNICZI A, MOHN C, VACAS I, & CEBRAL E, E. J., FRIEDMAN S, RETTORI V, BOYER PM 2002. Effect of nutritional stress on the hypothalamo-pituitary-gonadal axis in the growing male rat. Neuroimmunomodulation, 10, 153–162.

CONN, M. 1986. The molecular basis of gonadotropinreleasing hormone action. . EndocrRev 7, 3.

CONN, P. M., ROGERS, D. C., SEAY, S.G., STALEY, D. 1984. Activationofluteinizing hormone release from pituitary cells by polycations. Endocrinology 115 1913-1917.

CONN PM., STALEY D., HARRIS C., ANDREWS WV., GOROSPE WC., MCARDLE CA., HUCKLE WR. & J., H. 1986. MECHANISM OF ACTION OF GONADOTROPIN RELEASING HORMONE. Ann. Rev. Physiol, 48, 495-513.

CURTIS, K. K., SARANTOPOULOS, J., NORTHFELT, D. W., WEISS, G. J., BARNHART, K. M., WHISNANT, J. K., LEUSCHNER, C., ALILA, H., BORAD, M. J. & RAMANATHAN, R. K. 2014. Novel LHRH-receptor-targeted cytolytic peptide, EP-100: first-in-human phase I study in patients with advanced LHRH-receptor-expressing solid tumors. Cancer Chemotherapy and Pharmacology, 73, 931-941.

DE SOUZA MJ, L. D., VANHEEST JL, SCHEID JL, WEST SL, WILLIAMS NI 2007. Severity of energy-related menstrual disturbances increases in proportion to indices of energy conservation in exercising women. Fertility and Sterility, 88 971–975.

What About Multi-Part Projects?

We’ve got it—each section delivered on time, cohesive and high-quality. We’ll manage the whole journey for you.

DENG, X., QIU, Q., MA, K., HUANG, W. & QIAN, H. 2015. Synthesis and in vitro anti-cancer evaluation of luteinizing hormone-releasing hormone-conjugated peptide. Amino Acids, 47, 2359-66.

DERMAN SG., MCCLAMROCK HD. & EY., A. Regulation of the Pituitary Response to GonadotropinReleasing Hormone. Gynecology & Obstetrics. 2004 ed.: Lippincott Williams & Wilkins.

DHANDAPANI, K. M., BRANN, D.W., 2000. The role of glutamate and nitric oxide in the reproductive neuroendocrine system. . Biochem. Cell Biol., 78, 165–179.

ENGEL J, E. G., PINSKI J, SCHALLY A V. 2012. AEZS-108 : a targeted cytotoxic analog of LHRH for the treatment of cancers positive for LHRH receptors. Expert Opin Investig Drugs, 21, 891–9.

ERALY SA, M. P. 1995. Regulation of gonadotropin-releasing hormone transcription by protein kinase C is mediated by evolutionarily conserved promoter-proximal elements. . Mol Endocrinol 848–859.

Do You Adapt to International Rules?

Yes! UK, US, or Aussie standards—we’ll tailor your paper to fit your school’s norms perfectly.

EVANS, M. C. & ANDERSON, G. M. 2017. Neuroendocrine integration of nutritional signals on reproduction. J Mol Endocrinol, 58, R107-R128.

EVANS MC, R. M., MAYER C, BOEHM U & ANDERSON GM 2014. Evidence that insulin signalling in gonadotrophin-releasing hormone and kisspeptin neurones does not play an essential role in metabolic regulation of fertility in mice. Journal of Neuroendocrinology, 26, 468–479.

FARKAS I, V. C., SARVARI M, LIPOSITS Z. 2013. Ghrelin decreases firing activity of gonadotropin-releasing hormone (GnRH) neurons in an estrous cycle and endocannabinoid signaling dependent manner. PLoS ONE, 8.

FERNALD, R. D. & WHITE, A. R. B. 1999. Gonadotropin-Releasing Hormone Genes: Phylogeny, Structure, and Functions. Frontiers in Neuroendocrinology, 20, 224-240.

FERRIS H.A., S. M. A. 2006. Mechanisms for pulsatile regulation of the gonadotropin subunit genes by GNRH1. Biol. Reprod, 74, 993–998.

What does a complex assignment mean?

If your assignment needs a writer with some niche know-how, we call it complex. For these, we tap into our pool of narrow-field specialists, who charge a bit more than our standard writers. That means we might add up to 20% to your original order price. Subjects like finance, architecture, engineering, IT, chemistry, physics, and a few others fall into this bucket—you’ll see a little note about it under the discipline field when you’re filling out the form. If you pick “Other” as your discipline, our support team will take a look too. If they think it’s tricky, that same 20% bump might apply. We’ll keep you in the loop either way!

GAULT PM, M. S. 2003. Evidence That Gonadotropin-Releasing Hormone II Is Not a Physiological Regulator of Gonadotropin Secretion in Mammals. . J Neuroendocrinol 15, 831-839.

GONZALEZ–BARCENA D, V. B. M., GOMEZ–ORTA F, FUENTES GARCIA M, CARDENAS–CARNEJO I, GRAEF–SANCHEZ A, COMARU–SCHALLY AM, SCHALLY AV.  1994. Responses to the antagonistic analog of LH-RH (SB-75, Cetrorelix) in patients with benign prostatic hyperplasia and prostatic cancer. Prostate 24, 84-92.

GOODWIN, D., VARAMINI, P., SIMERSKA, P. & TOTH, I. 2015. Stability, permeability and growth-inhibitory properties of gonadotropin-releasing hormone liposaccharides. Pharm Res, 32, 1570-84.

GORE , A. & ROBERTS, J. 1997. Regulation of Gonadotropin-Releasing Hormone Gene Expression in vivo and in vitro. FRONTIERS IN NEUROENDOCRINOLOGY, 18, 209-245.

Who is my writer? How can I communicate with him/her?

Our writers come from all corners of the globe, and we’re picky about who we bring on board. They’ve passed tough tests in English and their subject areas, and we’ve checked their IDs to confirm they’ve got a master’s or PhD. Plus, we run training sessions on formatting and academic writing to keep their skills sharp. You’ll get to chat with your writer through a handy messenger on your personal order page. We’ll shoot you an email when new messages pop up, but it’s a good idea to swing by your page now and then so you don’t miss anything important from them.

HAHN, D. W., PHILLPS-PROBST, A., MCGUIRE, J.L., 1981. The need for controlling variables invloved in biologic assays of LHRH. In: IN: SCIARRA, J. E. (ed.) LHRH Peptides as Female and Male Contraceptives. Harper & Row. Philadelphia.

HARRISON, G. S., WIERMAN, M. E., NETT, T. M. & GLODE, L. M. 2004. Gonadotropin-releasing hormone and its receptor in normal and malignant cells. Endocr Relat Cancer, 11, 725-48.

HAYES FJ, T. A., MARTIN KA, HALL JE. 1998. Use of a gonadotropinreleasing hormone antagonist as a physiologic probe of polycystic ovary syndrome: Assessment of neuroendocrine and androgen dynamics. J Clin Endocrinol Metab 83, 2343-2349.

HERBISON, A. E. 1998. Multimodal influence of estrogen upon gonadotropin-releasing hormone neurons. Endocr. Rev. , 19, 302-330.

HERBISON, A. E., PAPE, J.R., 2001. Newevidence for estrogen receptors in gonadotropin-releasing hormone neurons. Front Neuroendocrinol. , 22, 292-308.

HISLOP JN, MADZIVA MT, EVEREST HM, HARDING T, UNEY JB, WILLARS GB, MILLAR RP, TROSKIE BE, DAVIDSON JS & CA., M. 2000. Desensitization and Internalization of Human and XenopusGonadotropin-Releasing Hormone Receptors Expressed in αT4 Pituitary Cells Using Recombinant Adenovirus. Endocrinology, 141, 4564-4575.

HONG IS, C. A., LEUNG PC. 2008. Gonadotropin-releasing hormones I and II induce apoptosis in human granulosa cells. . J Clin Endocrinol Metab 93, 3179 –3185.

HSUEH, A. J. W. & JONES, A. P. B. C. 1983. Gonadotropin Releasing Hormone: Extrapituitary Actions and Paracrine Control Mechanisms. Ann. Rev. Physiol., 45, 83-94.

HUANG W, A.-M. M., HORTON TH & LEVINE JE 2008 Fasting induced suppression of LH secretion does not require activation of ATP-sensitive potassium channels. . American Journal of Physiology: Endocrinology and Metabolism, 295, E1439–E1446.

IQBAL, J., SHAHNAZ, G., PERERA, G., HINTZEN, F., SARTI, F. & BERNKOP-SCHNÜRCH, A. 2012. Thiolated chitosan: Development and in vivo evaluation of an oral delivery system for leuprolide. European Journal of Pharmaceutics and Biopharmaceutics, 80, 95-102.

J. WAXMAN, A. M., W.F. HENDRY, H.N. WHITFIELD, G.M. BESSER, R.C. TIPTAFT, A.M. PARIS, R.T. OLIVER 1985. Importance of early tumour exacerbation in patients treated with long acting analogues of gonadotrophin releasing hormone for advanced prostatic cancer. British medical journal (Clinical research ed.), , 291 1387-1388.

J.E. RIVIER, J. P., C.L. RIVIER, M. PERRIN, A. CORRIGAN, W.A. HOOK, R.P. SIRAGANIAN, W.W. VALE, 1986. New effective gonadotropin releasing hormone antagonists with minimal potency for histamine release in vitro. Journal of medicinal chemistry, 29 1846-1851.

KAKAR SS., MUSGROVE LC. & DC., D. 1992. Cloning, sequencing and expression of human gonadotropin releasing hormone (GnRH) receptor. . Biochem Biophys Res Commun, 189.

KALRA, S. P. 1993. Mandatory neuropeptide-steroid signaling for the preovulatory luteinizing hormone-releasing hormone discharge. Endocr. Rev., 14 507–538.

KARTEN, M., RIVIER, J. 1986a. Overview of GnRH analog development. Endocrine Reviews, In press.

KARTEN, M. J., RIVIER, J.E., 1986b. Gonadotropin-releasing hormone analog design. Structure-function studies toward the development of agonists and antagonists: rationale and perspective. Endocr. Rev., 7, 44–66.

KENEALY, B. P. & TERASAWA, E. 2011. Rapid direct action of estradiol in GnRH neurons: findings and implications. Front Endocrinol (Lausanne), 2, 106.

KOTTLER ML, L. F., BERGAMETTI F, COMMERCON P, SOUCHIER C AND COUNIS R 1995. Subregional mapping of the human gonadotropin‐releasing hormone receptor (GnRH‐R) gene to 4q between markers D4S392 and D4S409. Hum Genet  96, 477–480.

KOVACS, M. & SCHALLY, A. V. 2001. Comparison of mechanisms of action of luteinizing hormone-releasing hormone (LHRH) antagonist cetrorelix and LHRH agonist triptorelin on the gene expression of pituitary LHRH receptors in rats. Proc Natl Acad Sci U S A, 98, 12197-202.

KRSMANOVIC LZ, S. S., MERELLI F, DUFOUR SM, VIRMANI MA, CATT KJ. 1992. Calcium signaling and episodic secretion of gonadotropin-releasing hormone in hypothalamic neurons. . Proc Natl Acad Sci USA 89, 8462–8466.

LA., R. & TM., N. 2005. Pituitary gonadotropin-releasing hormone(GnRH) receptor: Structure, distribution and regulation of expression. Animal Reproduction Science, 88, 57-74.

LAIMOU, D., KATSILA, T., MATSOUKAS, J., SCHALLY, A., GKOUNTELIAS, K., LIAPAKIS, G., TAMVAKOPOULOS, C. & TSELIOS, T. 2012. Rationally designed cyclic analogues of luteinizing hormone-releasing hormone: enhanced enzymatic stability and biological properties. Eur J Med Chem, 58, 237-47.

LAIMOU, D. K., KATSARA, M., MATSOUKAS, M.T.I., APOSTOLOPOULOS, V., TROGANIS, A.N., TSELIOS, T.V., 2010. Structural elucidation of Leuprolide and its analogues in solution: insight into their bioactive conformation. Amino Acids, 39, 1147–1160.

LI M, T. Z., ZHANG Y, LV S, LI Q, CHEN X. 2015. Targeted delivery of cisplatin by LHRH-peptide conjugated dextran nanoparticles suppresses breast cancer growth and metastasis. Acta Biomater, 18, 132–43.

LI, S., PELLETIER, G., 1992. Role of dopamine in the regulation of gonadotropin-releasing hormone in the male rat brain as studied by in situ hybridization. Endocrinology, 131, 395–399.

MAGGI, R., CARIBONI, A. M., MARELLI, M. M., MORETTI, R. M., ANDRE, V., MARZAGALLI, M. & LIMONTA, P. 2016. GnRH and GnRH receptors in the pathophysiology of the human female reproductive system. Hum Reprod Update, 22.

MARQUES P, S. K., ROZARIO KS, ET AL 2000. Physiology of Gnrh and Gonadotropin Secretion. In: DE GROOT LJ, C. G., DUNGAN K, ET AL (ed.) Endotext. Internet: South Dartmouth (MA): MDText.com, Inc.

MARSHALL JC, D. A., HAISENLEDER DJ, GRIFFIN ML, KELCH RP. 1993. GnRH pulses–the regulators of human reproduction. Transactions of the American Clinical and Climatological Association, 104, 31-46.

MERRIFIELD, R. B. 1966. The instrument for automated synthesis of peptides. Anal. Chem., 38.

MILLAR, R. P. 2005. GnRHs and GnRH receptors. Anim Reprod Sci, 88, 5-28.

MILLAR RP 2003. GnRH II and type II GnRH receptors. Trends Endocrinol Metab, 14, 35 –43.

MILLER, L. A., GIONFRIDDO, J. P., FAGERSTONE, K. A., RHYAN, J. C. & KILLIAN, G. J. 2008. ORIGINAL ARTICLE: The Single-Shot GnRH Immunocontraceptive Vaccine (GonaCon™) in White-Tailed Deer: Comparison of Several GnRH Preparations. American Journal of Reproductive Immunology, 60, 214-223.

MIYAMOTO K, H. Y. 1984. Identification of the second gonadotropin-releasing hormone in chicken hypothalamus: evidence that gonadotropin secretion is probably controlled by two distinct gonadotropin-releasing hormones in avian species. Proc Natl Acad Sci USA, 81, 3874-3878.

MOENTER, S. M., CARATY, A., LOCATELLI, A., KARSCH, F.J., 1991. Pattern of gonadotropin-releasing hormone (GnRH) secretion leading up to ovulation in the ewe: existence of a preovulatory GnRH surge. Endocrinology, 129, 1175–1182.

MONTAGNANI MARELLI, M., MORETTI, R. M., MAI, S., PROCACCI, P., LIMONTA, P. 2007. Gonadotropin-releasing hormone agonists reduce the migratory and the invasive behavior of androgen-independent prostate cancer cells by interfering with the activity of IGF-I. International Journal of Oncology 30, 261-271.

MORADI, S. V., VARAMINI, P., STEYN, F. & TOTH, I. 2015. In vivo pharmacological evaluation of a lactose-conjugated luteinizing hormone releasing hormone analogue. Int J Pharm, 495, 106-11.

NELSON, S. B., ERALY, S. A. & MELLON, P. L. 1998. The GnRH promoter: target of transcription factors, hormones, and signaling pathways. Mol. Cell. Endocrinol, 140, 151-155.

NUNEMAKER, C. S., DEFAZIO, R.A., MOENTER, S.M., 2002. Estradiol-sensitive afferents modulate long-term episodic firing patterns of GnRH neurons. Endocrinology 143, 2284–2292.

OKUBO K, N. Y. 2008. Structural and functional evolution of gonadotropin-releasing hormone in vertebrates. Acta Physiol (Oxf) 193, 3-15.

PADULA, A. M. 2005. GnRH analogues–agonists and antagonists. Anim Reprod Sci, 88, 115-26.

PARBORELL F, I. G., CELIN AR, TESONE M. 2008. Regulation of ovarian angiogenesis and apoptosis by GnRH-I analogs. Mol Reprod Dev, 75, 623–631.

PARKINSON, R. J., SIMMS, M. S., BROOME, P., HUMPHREYS, J. E. & BISHOP, M. C. 2004. A Vaccination Strategy for the Long-Term Suppression of Androgens in Advanced Prostate Cancer. European Urology, 45, 171-175.

PERRETT, R. M. & MCARDLE, C. A. 2013. Molecular mechanisms of gonadotropin-releasing hormone signaling: integrating cyclic nucleotides into the network. Front Endocrinol (Lausanne), 4, 180.

PINILLA L, A. E., DIEGUEZ C, MILLAR RP, TENA-SEMPERE M. 2012. Kisspeptins and reproduction: physiological roles and regulatory mechanisms. Physiol Rev, 3, 1235 –1316.

PINSKI J, L. N., HALMOS G, GROOT K, JUNGWIRTH A, VADILLO–BUENFIL M, KAKAR SS, SCHALLY AV. 1996. Chronic administration of luteinizing hormone-releasing hormone (LHRH) antagonist Cetrorelix decreases gonadotrope responsiveness and pituitary LHRH receptor messenger ribonucleic acid levels in rats. Endocrinology, 137, 3430-3436.

RISSMAN, E. F., LI, X., KING, J. A. & MILLA, R. P. 1997. Behavioral Regulation of Gonadotropin-Releasing Hormone Production. Brain Research Bulletin, 44, 459-464.

ROBERTSON, J. F. R. & BLAMEY, R. W. 2003. The use of gonadotrophin-releasing hormone (GnRH) agonists in early and advanced breast cancer in pre- and perimenopausal women. European Journal of Cancer, 39, 861-869.

ROMETO AM, K. S., VOYTKO ML, RANCE NE. 2007. Hypertrophy and increased kisspeptin gene expression in the hypothalamic infundibular nucleus of postmenopausal women and ovariectomized monkeys. J Clin Endocrinol Metab, 92, 2744-2750.

SAVOY-MOORE RT, S. K. 1987. Several GnRH stimulation frequencies differentially release FSH and LH from isolated, perfused rat anterior pituitary cells. Adv Exp Med Biol, 219, 641–645.

SCHALLY, A. V. 1999. Luteinizing hormone-releasing hormone analogs: their impact on the control of tumorigenesis. Peptides, 20, 1247-1262.

SCHALLY AV 2000. Use of GnRH in preference to LH-RH terminology in scientific papers. . Hum Reprod Update, 15, 2059-2061.

SCHALLY AV, N. A. 2004. Chemotherapy targeted to cancers through tumoral hormone receptors. Trends Endocrin Met, 15, 300-310.

SEALFON, S., WEINSTEIN, H., MILLAR, R., 1997. Molecular mechanisms of ligand interaction with the gonadotropinreleasing hormone receptor. Endocr. Rev. , 18, 180–205.

SHAM S. KAKAR & JENNES, L. 1995. Expression of gonadotropin-releasing hormone and gonadotropin-releasing hormone receptor mRNAs in various non-reproductive human tissues. Cancer Letters, 98, 57-62.

SKINNER, D. C., CARATY, A., ALLINGHAM, R. 2001. Unmasking the progesterone receptor in the preoptic area and hypothalamus of the ewe: no colocalization with gonadotropin-releasing neurons. Endocrinology, 142, 573-579.

SKORUPSKAITE, K., GEORGE, J. T. & ANDERSON, R. A. 2014. The kisspeptin-GnRH pathway in human reproductive health and disease. Hum Reprod Update, 20, 485-500.

THOMPSON, I. R. & KAISER, U. B. 2014. GnRH pulse frequency-dependent differential regulation of LH and FSH gene expression. Mol Cell Endocrinol, 385, 28-35.

TILBROOK AJ1, C. I. 1995. Negative feedback regulation of the secretion and actions of GnRH in male ruminants. J Reprod Fertil Suppl., 49, 297-306.

TILBROOK, A. J. & CLARKE, I. J. 1995. Negative feedback regulation of the secretion and actions of GnRH in male ruminants. J Reprod Fertil Suppl, 49, 297-306.

TILBROOK, A. J., TURNER, A.I., CLARKE, I.J., 2002. . Stress and reproduction: central mechanisms and sex differences in non-rodent species. . Stress, 5, 83–100.

TSUTSUMI M, Z. W., MILLAR RP, MELLON PL, ROBERTS JL, FLANAGAN CA, DONG K, GILLO B AND SEALFON SC 1992. Cloning and Functional Expression of a Mouse Gonadotropin-Releasing Hormone Receptor. Molecular Endocrinology, 1163-1169.

VARAMINI, P., MANSFELD, F. M., GIDDAM, A. K., STEYN, F. & TOTH, I. 2017. New gonadotropin-releasing hormone glycolipids with direct antiproliferative activity and gonadotropin-releasing potency. Int J Pharm, 521, 327-336.

VICKERY, B. H. 1986. Comparison of the potential for therapeutic utilities with gonadotropinreleasing hormone agonists and antagonists. Endocrine reviews, 7, 115-124.

WETSEL WC, E. S., WHYTE DB, MELLON PL. 1993. Regulation of gonadotropin-releasing hormone by protein kinase-Aand -C in immortalized hypothalamic neurons. . Endocrinology  132, 2360–2370.

WHITE SA, B. C. 1994. A second gene for gonadotropin-releasing hormone: cDNA and expression pattern in the brain. Proc Natl Acad Sci USA, 91, 1423-1427.

WILSON AC, M. S., BOWEN RL, ATWOOD CS. 2007. Leuprolide acetate: a drug of diverse clinical applications. Expert Opin. Investig. Drugs, 16, 1-13.

YANG-FENG TERESA L., SEEBURG PETER H. & UTA., F. 1985. Human Luteinizing Hormone-Releasing Hormone Gene (LHRH) Is Located on Short Arm of Chromosome 8 (Region 8p11.2 , p21) Somatic Cell and Molecular Genetics, 12, 95-100.

ZVI NAOR, O. B., RONY SEGER, 2000. Activation of MAPK Cascades by G-protein-coupled Receptors: The Case of Gonadotropin-releasing Hormone Receptor,. Trends in Endocrinology & Metabolism, 11, 91-99.

Tags: Affordable Online College Homework, Cheap essay writer Australia, Pay someone to write my paper, Research Essay Help UK

Order|Paper Discounts

Why Choose Essay Bishops?

You Want The Best Grades and That’s What We Deliver

Top Essay Writers

Our top essay writers are handpicked for their degree qualification, talent and freelance know-how. Each one brings deep expertise in their chosen subjects and a solid track record in academic writing.

Affordable Prices

We offer the lowest possible pricing for each research paper while still providing the best writers;no compromise on quality. Our costs are fair and reasonable to college students compared to other custom writing services.

100% Plagiarism-Free

You’ll never get a paper from us with plagiarism or that robotic AI feel. We carefully research, write, cite and check every final draft before sending it your way.