Open Access

Hormone supply to the pituitary gland: A comprehensive investigation of female‑related tumors (Review)

  • Authors:
    • Wenxiu Tian
    • Huimin Qi
    • Zhimei Wang
    • Sen Qiao
    • Ping Wang
    • Junhong Dong
    • Hongmei Wang
  • View Affiliations

  • Published online on: August 9, 2022     https://doi.org/10.3892/ijmm.2022.5178
  • Article Number: 122
  • Copyright: © Tian et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The hypothalamus acts on the pituitary gland after signal integration, thus regulating various physiological functions of the body. The pituitary gland includes the adenohypophysis and neurohypophysis, which differ in structure and function. The hypothalamus‑hypophysis axis controls the secretion of adenohypophyseal hormones through the pituitary portal vein system. Thyroid‑stimulating hormone, adrenocorticotropic hormone, gonadotropin, growth hormone (GH), and prolactin (PRL) are secreted by the adenohypophysis and regulate the functions of the body in physiological and pathological conditions. The aim of this review was to summarize the functions of female‑associated hormones (GH, PRL, luteinizing hormone, and follicle‑stimulating hormone) in tumors. Their pathophysiology was described and the mechanisms underlying female hormone‑related diseases were investigated.

1. Introduction

To systemically study the association between pituitary hormones and female-related tumors, a PubMed search was performed using the key terms 'growth hormone', 'prolactin', 'luteinizing hormone', 'follicle-stimulating hormone' or 'female', including articles from 2017 to 2021, with some exceptions from the last 10 years. The search strategy included research articles and reviews. In total, 158 studies were included in the present review. The pituitary gland is the most important endocrine gland in the human body, controlling the secretion of key hormones involved in metabolism, growth, development, and reproduction. Six main hormones are secreted by adenohypophyseal cells: growth hormone (GH), prolactin (PRL), thyroid-stimulating hormone (TSH), gonadotropin [luteinizing hormone (LH) and follicle-stimulating hormone (FSH)], and adrenocorticotropic hormone. In the present review, the association between the GH, gonadotropin-related hormones, and PRL, and the occurrence of associated diseases was summarized, with a focus on tumor development.

2. Growth hormone

The regulation of hypothalamic GH secretion is mediated by the growth hormone-releasing hormone (GHRH), somatostatin (SST), and ghrelin (1). GHRH promotes GH release by interacting with the growth hormone-releasing hormone receptor (GHRHR) in the somatotroph cells of the adenohypophysis through the cyclic adenosine monophosphate (cAMP)-mediated signal transduction pathway (2). SST inhibits GH secretion by binding to SST receptors (2,3). In addition, ghrelin, a hormone-releasing peptide, promotes GH secretion by activating the growth hormone secretagogue receptor (GHS-R) in the hypothalamus and pituitary (3). Growth hormone receptors (GHRs) are widely distributed through the body, including in bones, muscles, heart, brain, kidney, and fat cells (4). GH secretion may vary with age, sex, and nutritional status, peaking at a young age due to increased estrogen or testosterone concentrations (4-6).

Effects of GH on growth and development. GH misregulation is linked to several diseases. Growth hormone deficiency (GHD) incidence is highest in children. It has been reported that children with GHD after birth are often characterized by slow growth and short stature (7,8). Being a positive modulator of growth, GH is used to treat diseases characterized by short statures, such as short stature homeobox (SHOX) deficiency (linked to SHOX gene mutations), Turner syndrome, Prader-Willi syndrome, Noonan syndrome, and small for gestational age (SGA) (9-13). GH regulates human growth by modulating the metabolism of carbohydrates, proteins and lipids (14). Moreover, adults with GHD experience increased total and visceral body fat, low bone and muscle mass, reduced muscle strength, impaired anaerobic performance, poor cardiovascular status, and poor quality of life (15,16). Previous research has shown that GH acts directly in chondrocytes to promote differentiation and chondrocyte proliferation, by stimulating peripheral tissues via upregulation of the insulin-like growth factor-1 (IGF-1), especially in the liver (17). In adults, excessive growth hormone secretion and increased IGF-1 concentration may lead to acromegaly. In children, excessive secretion of GH and IGF-1 leads to gigantism (18,19). Therefore, accurate regulation of GH is crucial to modulate human growth and development at all ages.

Additional functions of GH

Some evidence shows a close association between the neuroendocrine and the immune systems. In vitro and in vivo studies have demonstrated that GH is involved in immune response, by regulating the proliferation and activity of immune cells, namely the proliferation of leukocyte subsets with expression of GHRs (20). GH-activated protein kinase C (PKC) and ERK-activated hormone-sensitive lipase (HSL) regulate lipolysis and improve sleep efficiency (21,22). Furthermore, it is likely that GH exerts important roles in ovulation and fertility of women (23). Polycystic ovary syndrome (PCOS) is the most common female endocrine disorder, affecting 5-10% of women at reproductive age. Oxidative stress (OS) plays a central role in the pathophysiology of PCOS and other diseases. GH reduces OS-induced apoptosis by activating the PI3K/Akt signaling pathway, which may provide a theoretical basis for PCOS treatment (24). Furthermore, GH administration has been associated with successful in vitro fertilization (IVF). Exogenous GH administration alleviates mitochondrial dysfunction and improves oocyte quality in women above 40, and in patients with poor ovarian response. Studies have shown that GH treatment during ovarian stimulation in young women who have failed IVF may increase IVF success rates by improving oocyte quality (25).

Research progress in GH and female GH-related tumors

The incidence of malignant tumors, including breast cancer, is major public health concern in Chinese women. A significant portion of cancers are hormone-dependent, and GH plays key roles in the proliferation and invasion of breast cancer cells (26). Human GH (hGH) promotes body growth via activation of distinct biological processes, including IGF-1 synthesis and secretion in the liver, and activation of JAK2/STAT5 and BRAF/MEK/ERK signaling pathways. GH triggers cell proliferation in breast cancer cells by increasing JAK2 and STAT5 gene expression. Conversely, silencing of GHRs blocks JAK2/STAT5 and BRAF/MEK/ERK signaling pathways, thereby inhibiting the growth of breast cancer cells (27,28). Clinicopathological studies of cases with breast cancer revealed that hGH expression was positively correlated with the presence of metastases, high clinical stage and HER2 positive lymph nodes. It has also been reported that elevated GH levels may induce malignant transformation of mammary epithelial cells, promote proliferation and metastasis via activation of epithelial-mesenchymal transition (EMT), through miR-183-96-182 cluster activation. During EMT, epithelial cells reduce their polarity and intercellular contacts, therefore acquiring migration and motility capacities, enabling them to invade adjacent tissues and distant tissues. EMT is triggered after cells receive microenvironmental signals and it is activated during the pathogenesis of cancer (29-31).

GH promotes the proliferation and invasion of breast cancer cells by promoting EMT. GH-mediated EMT in breast cancer may occur via two distinct mechanisms: i) GH may directly promote EMT, or ii) GH may promote the EMT of breast cancer cells by inducing microRNA clustering and inhibiting the expression of breast cancer metastasis suppressor 1-like (BRMS1L) (32,33). In addition, GH may promote the development of breast cancer cells by downregulating connexin E-cadherin expression and inducing the WNT signaling pathway (32,34). Abnormal secretion of GH is additionally associated with several human diseases. Previous research has revealed that hGH expression is increased in hepatocellular carcinoma (HCC) and is associated with poor survival outcomes in HCC patients. hGH secreted by HCC promotes invasion and cancer stem cell-like properties by inhibiting the expression of the tight junction component CLAUDIN-1. Thus, inhibiting hGH and hGHR signaling pathways may be a potential therapeutic approach to limit HCC progression and recurrence (35). The specific molecular mechanism is presented in Fig. 1.

Drug treatment of GH-related diseases

Excessive GH secretion is often surgically treated, followed by post-operative adjuvant drug therapy, which may include GHR antagonists, dopamine agonists (DAs), and SST receptor ligands. Pegvisomant is a recombinant GHR antagonist, similar to hGH, that binds and blocks GHR activity (36). Pegvisomant treatment reestablishes normal levels of IGF-1 in patients with acromegaly and relieves symptoms related to excess GH (37). Cabergoline (CAB) is a dopamine agonist that can be used in combination with pegvisomant. CAB is the first-line treatment for hyperprolactinemia, being additionally used to treat acromegaly (38,39). SST is a cyclic-released hormone that functions as an inhibitory peptide. SST receptor ligand drugs, such as octreotide, lanreotide, and pasireotide, are used in the clinic to treat diseases caused by the excessive release of GH. Octreotide and lanreotide are specific for the SST receptor somatostatin receptor 2 (SSTR2), a key regulator of GH secretion (40,41). Acromegaly is characterized by hypersecretion of GH and elevated levels of IGF-1. The reported leading cause of acromegaly is the presence of a GH-secreting pituitary neuroendocrine tumor (PitNET) or GH plus PRL-secreting mixed adenoma. Combination therapy including SST and dopamine inhibit GH and IGF-1 secretion by binding to SSTR2 and dopamine D2 receptor (D2R), thereby inhibiting the acral lesions caused by GH-secreting PitNETs (41-43).

3. Prolactin

PRL is a versatile hormone and serves a wide variety of physiological functions apart from lactation. PRL lactotrophs in the pituitary are released in a circadian pattern depending on the physiological state. PRL content in the human body varies slightly with sex; with women presenting higher levels than men during puberty (44,45). PRL further participates in immune regulation, by binding to PRL receptors (PRLRs) in the surface of monocytes, lymphocytes, and thymic epithelial cells (46,47). PRL secretion is controlled by hypothalamic PRL release factors and PRL release-inhibiting hormones, which include TSH-releasing hormone, vasoactive intestinal peptide, serotonin, PRL release-inhibiting hormone dopamine class material, growth hormone-releasing inhibitory hormone (GHIH), gamma-aminobutyric acid (GABA), and thyroid hormone, among others (48). In addition, PRL regulates luteal function via modulation of LH receptors in the ovaries (49).

Role of PRL in pregnancy

PRL belongs to the family of growth factors that regulate cellular and humoral immunity. During pregnancy, PRL is key for intrauterine homeostasis and normal embryo development. PRL levels in women who have suffered from miscarriage are significantly lower than in women with healthy pregnancies. Moreover, low levels of PRL are associated with low weight at birth and premature birth (50).

PRL is synthesized in the endometrium during embryo implantation, providing a suitable microenvironment for blastocyst implantation through PRLR, to improve the endometrial receptivity. Precise regulation of PRL concentration is key to promote growth and adhesion of the endometrial cells which are essential for embryo implantation and development (51). The space-temporal regulation of PRLR expression suggests that PRL regulates several functions related to mitosis and differentiation of ovarian tissue. PRL production is positively correlated with the degree of the decidualization of endometrial stromal cells (48). PRL secreted by decidua during pregnancy increases after embryo implantation, reaching the peak around 20-25 weeks of pregnancy (52). PRLR expression has further been detected in the placental trophoblast, and amniotic epithelial cells (53). Thus, PRL should be adequately added as hormone replacement therapy in premature infants (54).

In recent years, the association between human T helper cells (Th1/Th2) in serum, lymphocytes, decidua, and spontaneous abortion has attracted much attention. It is maintained that Th1 cells inhibit embryo development. In contrast, Th2 cells play key roles in maintaining a normal pregnancy. PRL regulates the Th1/Th2 balance, particularly acting on Th1 cells. In addition, PRL decreases natural killer (NK) cells and Th1 activity. PRL and PRLR are abundantly expressed in uterine decidual tissue in early pregnancy (55-57). PRL further stimulates the expression of LH/placental secretory chorionic gonadotropin receptor (CG receptor) in the corpus luteum after fertilization. Moreover, PRLR, coordinates with FSH and LH, thereby regulating cAMP and progesterone synthesis, both of which are essential during pregnancy (58-60). Based on these studies, PRL may regulate the local microenvironment of uterine decidua that controls the synthesis and secretion levels of hormones and factors by autocrine and paracrine pathways. Endogenous or exogenous factors blocking PRL synthesis and secretion may affect the expression of LH/CG receptors in the pregnant corpus luteum of the ovary, which may result in miscarriage. In conclusion, PRL levels during early pregnancy are essential to normal embryo development, acting via luteal maintenance, immune function regulation, and decidual development.

Research progress in PRL and female PRL-related tumors

PRL is an essential growth factor with direct influence on the division and proliferation of somatic cells. Particularly, PRL plays a key role in regulating breast development and terminal differentiation of breast epithelial cells. Epidemiological research has revealed that elevated serum PRL levels are associated with increased risk of breast cancer. PRL and PRLR overexpression is observed in several tumors, including breast cancer, and is associated with increased cell proliferation, reduced apoptosis, and a shorter cell cycle (61).

The association between PRL and breast cancer has been thoroughly studied. Li et al determined that the PRL/p-STAT5 pathway induced HER2+ mammary tumorigenesis through oxytocin receptor (62). Another study revealed that two small molecule inhibitors, SMI-1 and SMI-6, abrogated PRL-induced HER2+ breast cancer cell invasion and malignant lymphocyte proliferation by binding the extracellular domain of PRLR (63). Moreover, PRL synergizes with canonical WNT signals and KRAS activation to drive the development of ER+ mammary tumors, by activating the Notch signaling pathway (64,65). MacDonald et al reported that PRL-inducible EDD E3 ubiquitin ligase promotes TORC1 signaling, anti-apoptotic protein expression, and drug resistance in mammary tumors (66). Chen et al used hybridoma technology to generate an anti-idiotypic antibody named H53. H53 exhibits biological activity against breast cancer, suggesting that an internal image with anti-idiotypic antibodies may be a candidate strategy to develop PRLR/GHR dual-function antagonists for breast cancer therapy (67). PRL has further been shown to promote pancreatic ductal adenocarcinoma, which, notably, can be reversed by antipsychotic diphenylbutylpiperidines (68). Moreover, blood PRL levels are reduced in rats with diabetes or obesity. In the clinical setting, it has been observed that low circulating levels of PRL are correlated with increased prevalence of diabetes and a higher risk of metabolic alterations, by increasing β-cell loss and pancreatic inflammation (69). PRL misregulation is also associated with glioma, uterine, prostate, renal cell, and pituitary tumors. In uterine cancer, blocking PRLR induces FOXO3a/EIF-4EBP1-mediated cell death (70). In glioma, the therapeutic block of PRLR enhances the response of tumor cells to chemotherapy (71). PRL promotes the expression of inducible nitric oxide synthase (iNOS) in glioma cells, thereby promoting the production of nitric oxide (NO). Furthermore, PRL leads to the accumulation of NO and the expression of iNOS by enhancing IFN-γ. In the prostate, PRL drives tumorigenesis by regulating STAT5a/b (72). The specific molecular mechanism is presented in Fig. 2.

Additional functions of PRL

PRL is involved in several biological processes that include immune response, neuroprotection, and metabolic regulation. PRL participates in immune regulation by interacting with PRLR on the surface of immune cells such as leukocytes, macrophages, lymphocytes, among others (46). PRL misregulation is associated with increased risk of multiple sclerosis and rheumatoid arthritis by regulating the number of circulating lymphocytes (57). PRL has been reported to exert neuroprotective functions. For instance, in a rat glutamate (Glu) injury model, PRL maintained the function of neuronal mitochondria by promoting intracellular calcium release and NF-κB. PRL further regulates nerve regeneration of non-transferrin bound iron (NTBI) by preventing calcium overload (73,74). Finally, PRL maintains physiological homeostasis through the regulation of the level of insulin, liver-related enzyme activity, and intestinal Ca2+ absorption (75-77).

Treatment of PRL-related diseases

Excessive secretion of PRL may lead to hyperprolactinemia, which is frequent in hermaphroditism. DAs are used as first-line treatment. Most commonly used drugs are CAB, bromocriptine (BRC), and pergolide (PER) (78-80), all of which act on dopamine receptors in the lactating cell membrane of the pituitary gland to reduce serum levels of PRL (81), thereby relieving the clinical symptoms of patients, and recovering the menstruation and fertility of women. Antipsychotics also increase the level of PRL. Among them, amisulpride, risperidone, and paliperidone significantly increase the levels of PRL, even at low doses (82). PRL dysfunction inhibits the male reproductive system and reduces semen quality (83). The antipsychotic drug aripiprazole (ARI) decreases serum PRL levels in children and adolescents (84). With the increasing research on the molecular and cellular biology of the pituitary, different target genes and gene transfer methods are under study as therapeutic candidates against PitNET. In the future, gene therapy will likely focus on the treatment of invasive PitNET and residual drug resistance diseases with local invasion following surgery (85). Gene therapy will undoubtedly enlighten the future clinical treatment of infections caused by abnormal PRL secretion.

4. Gonadotropin

Gonadotropin-releasing hormone (GnRH) is a heterodimeric glycoprotein composed of an α subunit and a β subunit (86-88). Gonadotropin, a member of the glycoprotein hormone family, consists of FSH and LH. There are several subtypes of FSH generated by post-translational modifications, all of which affect FSH half-life through the change of carbohydrate pattern (86). FSH participates in follicular development and regulates follicular secretion, maturation, and atresia by modulating granulosa cells. Late follicular stages may induce LH receptor (LHR) synthesis by granulosa cells, thus supporting LHR interaction with LH, resulting in the production of a luteinizing follicle (87). In women, LH stimulates the secretion of estrogen by theca cells, promotes ovulation, and maintains the physiological role of luteinization. In addition, inhibition of the hypothalamus is performed by inhibin A to regulate the menstrual cycle of women. GnRH is a therapeutic drug that stimulates the release of inhibin A, enhances the activity of LH, selectively inhibits the synthesis and secretion of FSH, and promotes the selection of dominant follicles (DF), thereby regulating reproductive function (89-92). Duan et al described four luteinizing hormone-choriogonadotropin receptor (LHCGR) structures using cryo-electron microscopy (cryo-EM): Two of the structures were the wild-type receptor in the inactive or in the active states; and the other two structures were the constitutively active mutated receptor. Research has revealed a unique mechanism of receptor activation, providing a rationale for drug discovery in endocrine-related diseases (93,94).

Neuromodulation of gonadotropin

A new action factor, the neuropeptide kisspeptin, which acts upstream of GnRH, has attracted the attention of the scientific community in recent years (95). Other neuropeptides (gonadotropin inhibitory hormone/radiofrequency amide-related peptide and other members of the radiofrequency amide peptide superfamily), and various non-peptide neurotransmitters (glial fibrillary acidic protein, dopamine, neuron-derived neurotrophic factor, and serotonin), also function as regulators of GnRH secretion and synthesis, as well as of LH and FSH secretion (95-98). Kisspeptin/neurokinin B/dynorphin (KNDy) neuron is a GnRH pulse generator that maintains gonadotropin levels and follicular development (99). Nuclear receptors, such as the steroid generation factor 1 and liver receptor homolog 1 (LRH-1), regulate progesterone receptors to control FSH and LH. These are also critical regulators of steroid generation, cell proliferation and migration, and cytoskeleton remodeling (100). Concurrently, makorin ring-finger protein 3 (MKRN3) inhibits the reproductive axis through its role in kisspeptin-expressing neurons involved in a MKRN3-guided ubiquitination mediation mechanism (101,102). A previous study revealed that a high-fat diet (HFD) intake by parents may also be a risk factor for prostate hyperplasia and cancer at advanced ages, due to a negative impact on the reproductive system of male offspring (102,103). Thus, it is advised that epidemiological and clinical research on semen quality must include male offspring of overweight and/or obese parents (103,104).

Effect of FSH on reproductive development

Gonadotropin plays a key role in reproduction, and the evaluation of FSH and LH is an index of ovarian reserve. There are several recombinant preparations used to treat gonadotropin deficiency and gonadal dysfunction. Abnormal FSH may lead to abnormal ovarian function, such as follicular development disorder and abnormal follicular atresia (105,106). The combination of urinary follicle-stimulating hormone (uFSH) and recombinant follicle-stimulating hormone (rFSH) improves ovarian stimulation (107). In addition, GnRH upregulates the anti-Mullerian hormone (AMH), which leads to ovulation dysfunction and androgen elevation by causing abnormal follicular growth and abnormal secretion of granulosa cells (108). Furthermore, GnRH affects bone mineral density in postmenopausal women by regulating LH and FSH levels (109). Postmenopausal osteoporosis is a risk factor for bone fracture. It was initially proposed that estrogen decay was responsible for the onset of postmenopausal osteoporosis. However, recent studies have shown that FSH, which increases along with estrogen decreasing, acts on FSH receptor (FSHR) in osteoclasts. FSH promotes osteoclast formation and accelerates bone loss by enhancing the expression of receptor activator of nuclear factor-κB (RANK). This suggests that FSH, along with estrogen, may be linked to the risk of osteoporosis in postmenopausal women (110,111). In addition, LH and FSH are distinctly regulated. For example, LH and FSH reduce nicotine-induced oocyte autophagy in different manners. LH reduces nicotine-induced autophagy by restoring the phosphorylation of adenosine 5′-monophosphate-activated protein kinase α-1. By contrast, FSH reduces autophagy by downregulating the phosphorylation of forkhead box O1 (FoxO1) and light chain 3 (LC3)-II (112,113). Paradoxically, FSH upregulates Beclin1 through the PI3K/JNK/c-Jun pathway to accelerate the degradation of lipid droplets in mammalian follicular granulosa cells, thus enhancing autophagy. FSH affects follicular development, and participates in the regulation of lipid metabolism (such as cholesterol and fat), regulates bone density, and is associated with the onset of cardiovascular diseases, and the occurrence of breast cancer (112). FSH activates liver cholesterol biosynthesis and decreases serum cholesterol through the Gi2α/β-inhibin-2/Akt pathway during menopause (114). Elderly postmenopausal women with higher FSH levels have higher bone marrow obesity rates, lower bone density, fat, and lean meat mass than elderly postmenopausal women with lower FSH levels (115,116). It is also suggested that FSH regulates bone formation and regeneration, and that its dysregulation enhances joint inflammation in rheumatoid arthritis. Mechanistically, FSH triggers pathogenesis through the regulation of activin A or inhibin B in the respiratory system (Fig. 3) (117,118).

Effects of LH on reproductive development

At present, research on LH function is mainly focused on the maturation and development of follicles and the recovery of the hematopoietic system (119,120). LH coordinates the process of oocyte meiosis via the activation of zinc finger protein 36 (ZFP36) expression in an EGFR-ERK1/2 dependent pathway (86,121). Moreover, LH participates in the homeostasis of hematopoietic stem cells (HSC). The lack of LHCGR has been revealed to accelerate the development of acute myeloid leukemia in mice (119). The LH blocker leuprorelin improves hematopoietic recovery after radiotherapy or chemotherapy, by protecting LHR-expressing HSCs (122,123). Additionally, LH is an inflammation modulator. During follicular maturation and development, the first responders to the LH wave are granulosa and theca cells, which produce steroids, prostaglandins, chemokines, and cytokines. These mediators activate ovarian cells and resident immune cells in the ovary, and further attract additional immune cells to the ovaries. These jointly regulate the proteolytic pathway to recombine the follicular matrix, destroy the basal layer of granulosa cells and promote the invasion of vascular endothelial cells (124,125). In mice, it was revealed that LH reduced the expression of the apoptosis-promoting protein Tap63, inhibited cisplatin-induced oocyte apoptosis, and protected the reproductive function of female mice following treatment (126). In addition, Li et al reported that p62 depletion in the pituitary impairs LH synthesis through mitochondrial OXPHOS signal transduction and leads to female infertility. This provided a new theoretical basis for studying female reproductive dysfunction in gonadotropic cells: The GnRH-p62-oxphos (ndufa2)-Ca2+/ATP-LH pathway (127). Notably, LH has been described as a therapeutic candidate in treating Alzheimer's disease (AD) and recovering from cognitive impairment. The increase in LH has been associated with increased risk of cognitive impairment and AD. Cognitive improvement after LH level reestablishment, and remission of AD symptoms after treatment with specific LHCGR blockers, have been observed (128,129). The precise molecular mechanism is revealed in Fig. 4.

Research progress on gonadotropin-related hormones and female-related tumors

In recent years, the expression of GnRH and its receptor have been found in a variety of gynecological tumors, including uterine fibroids, endometrial cancer, ovarian cancer, and breast cancer. Ovarian cancer is a common reproductive tumor in women. The most frequent subtype is epithelioid ovarian cancer (EOC), which can be further divided in serous ovarian cancer (SOC), endometrioid carcinoma, clear cell carcinoma (CCOC), mucinous carcinoma and other less diagnosed types (130,131).

Previous studies have shown that genetic, hormonal, and reproductive factors affect the incidence of ovarian cancer, but its etiology and pathogenesis remain unclear. The hypothalamus-pituitary ovary axis is an important neuroendocrine system that plays a crucial role in regulating the female reproductive system. Gonadotropin, as the key hub of this axis, plays an important role in ovarian cancer incidence (132). Tumor cell metabolic reprogramming and microenvironmental changes have been linked to the development of ovarian tumors (133-135). In this context, FSH and LH increase the synthesis of cyclooxygenase (COX)1 and COX2 through the PI3K/Akt signaling pathway and the production of prostaglandin E2 (PGE2), which itself is linked to disease progression (hypertension, diabetes, and dyslipidemia) (136-138). PEG2 promotes the progression of ovarian cancer by increasing the production of matrix metalloproteinase (MMP)2 and MMP9 (139,140). In addition, FSH promotes ovarian cancer progression by affecting the glycolysis process. For instance, FSH promotes glycolysis in ovarian cancer cells and the decrease of pH in the microenvironment by promoting the expression of pyruvate kinase M2 (PKM2) and phosphoglycerate kinase 1 (PGK1) (141,142). Therefore, gonadotropin may be involved in the progression of ovarian cancer by modulating tumor cell metabolism.

FSH affects the expression of progranulin (PGRN) in CCOC, through activation of the PI3K/Akt pathway via FSHR, thereby supporting the development of ovarian cancer. Nevertheless, FSH regulates different molecular mechanisms via PKC modulation in different subtypes of ovarian cancer including SOC (143). FSH increases the expression of Gankyrin through the PI3K/Akt pathway, and promotes the proliferation of ovarian cancer cells by regulating cyclin D1 (144). In addition, the specific expression of FSH in the reproductive system supports the hypothesis that FSH may be a candidate therapeutic target in ovarian cancer (145-147). LH also regulates the PI3K/Akt pathway by upregulating vascular endothelial growth factor (VEGF) and Slit2 (148). VEGF can be used as a gonadotropin to promote the progression of advanced ovarian cancer (149). In conclusion, gonadotropin is closely related to the occurrence and development of ovarian cancer, and may provide insights for targeted treatment.

Treatment of gonadotropin-related diseases

The treatment of gonadotropin-associated disorders is performed by gonadotropin-releasing hormone agonists (GnRH-a), gonadotropin-releasing hormone antagonists (GnRH ant), estrogen receptor modulators, aromatase inhibitors, and hormone analogs. GnRH-a promotes the release of gonadotropin. However, the long-term use of the GnRH-a leuprorelin results in decreased bone mineral density (150,151). The GnRH ant, cetrorelix inhibits the synthesis and secretion of gonadotropin by competing with GnRH receptor on the surface of pituitary cells to block GnRH secretion (152-154). In addition, clomiphene citrate, an estrogen receptor regulator, increases the secretion of GnRH in the hypothalamus by inhibiting the negative feedback regulation of estrogen. In addition to treating female ovulation dysfunction, clomiphene citrate also improves the levels of testosterone and GnRH in men (155-157). Aromatase inhibitors, which are estrogen receptor regulators, promote the release of GnRH by modulating estrogen levels. Furthermore, gonadotropin drugs are used to replace endogenous hormones. For example, human chorionic gonadotropin (hCG) is used as a LH substitute to induce ovulation (158). Hormone administration is often more effective than oral administration, however at the cost of the occurrence of side effects, such as increasing the risk of multiple births and ovarian hyperstimulation syndrome (OHSS).

5. Conclusions

In recent years, major progress in our knowledge concerning the mechanisms of action of the three pituitary hormones (GH, gonadotropin-related hormones, and PRL) and their roles in human health and diseases has advanced significantly, particularly their involvement in growth and development. GH promotes body growth and development, and its abnormal secretion is associated with a variety of diseases. GH is involved in breast cancer through dysregulation of the JAK2/STAT5 pathway and promotion of EMT. PRL is involved in distinct biological processes: It promotes milk secretion and gonad development, and is essential for metabolism, immunity, and fetal growth and development. In addition, PRL misregulation is linked to human diseases including breast and liver diseases, as well as autoimmune and endocrine diseases. PRL-based therapy is used to manage pain, particularly migraines in women. Moreover, PRL-based treatments are applied in the context of different cancers, including, but not limited to, pancreatic ductal carcinoma, glioma, breast cancer, uterine cancer, and prostate cancer. Dysregulation of LH and FSH hormones is associated with diseases of reproductive development in both men and women. In this context, an accurate FSH-LH balance is critical to seminiferous tubular development in early sexual maturation. The increase of FSH has been observed in primary amenorrhea, congenital ovarian hypoplasia, and primary hyper reproductive function. During the menstrual cycle, an increase in FSH is observed during the fertile period. Elevated FSH levels in the follicular phase are a marker of decreased ovarian function, which in turn leads to secondary decreases in ovarian estrogen and progesterone secretion. LH induces testosterone synthesis, thus supporting the production of mature sperm. Therefore, reduced LH levels are associated with a decrease in fertility, and ultimately with infertility.

The exact role of pituitary hormones in metabolic homeostasis remains to be fully elucidated, with recently described functions expanding further than the standard roles of these hormones. GH plays a unique biological role in various stages of human development through GHRs which are expressed in different organs. Lack of GH in childhood leads to slow growth and development, and to dwarfism in extreme cases. In adults, lack of GH leads to increased fat content and a high risk of cardiovascular disease. Moreover, GH impacts fertilization by affecting egg quality, and alleviates PCOS by reducing OS. An excessive gonadotropin level in adults is an indicator of premature ovarian failure, whereas its excessive levels confer a higher risk for bone loss and cognitive impairment during the perimenopausal period. Similarly, PRL functions are timely regulated. PRL promotes lactation in women, and its accurate regulation is fundamental for the development of healthy pregnancies. Thus, data indicate that these hormones play indispensable roles in different growth stages of the human body.

Overall, hormone regulation of the pituitary function is extremely complex and integrates multiple regulatory levels, ranging from subcellular to extracellular processes. Hormonal recombination agents are more effective than oral drugs. Although hormonal recombination agents are used to treat developmental abnormalities and reproductive disorders, the therapeutic effects come at the expense of several side effects, such as the increased risk of multiple births, in cases where gonadotropins are used to treat infertility. The use of hormonal therapy, using single or multiple agents requires further research, in order to limit the currently observed side effects of such treatment approaches. The present review integrated recent research results, aiming to provide new guidelines for future treatment strategies for clinical management and further drug development.

Availability of data and materials

Not applicable.

Authors' contributions

ZW performed the data collection. HQ performed the data curation. HW wrote the manuscript. WT and SQ prepared the figures. JD and PW revised the manuscript. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present review was financially supported by the National Natural Science Foundation of China (grant nos. 81602327 and 81500798) and the Funds for Zhishan Young Scholars (Southeast University; grant no. 2242021R41070).

Abbreviations:

AD

Alzheimer's disease

CAB

cabergoline

CCOC

clear cell carcinoma

COX

cyclooxygenase

DAs

dopamine agonists

EMT

epithelial-mesenchymal transition

FSH

follicle-stimulating hormone

FSHR

FSH receptor

GH

growth hormone

GHD

growth hormone deficiency

GHRs

growth hormone receptors

GHRH

growth hormone-releasing hormone

GnRH

gonadotropin-releasing hormone

GnRH-a

gonadotropin-releasing hormone agonists

GnRH ant

gonadotropin-releasing hormone antagonists

HCC

hepatocellular carcinoma

hGH

human GH

HSC

hematopoietic stem cells

HSL

hormone-sensitive lipase

IGF-1

insulin-like growth factor-1

iNOS

inducible nitric oxide synthase

IVF

in vitro fertilization

LC3

light chain 3

LH

luteinizing hormone

LHCGR

luteinizing hormone-choriogonadotropin receptor

LHR

LH receptor

MKRN3

makorin ring-finger protein 3

MMPs

matrix metalloproteinases

OS

oxidative stress

PCOS

polycystic ovary syndrome

PitNET

pituitary neuroendocrine tumor

PKC

protein kinase C

PRL

prolactin

PRLRs

PRL receptors

RANK

receptor activator of nuclear factor-κB

SGA

small for gestational age

SHOX

short stature homeobox

SOC

serous ovarian cancer

SST

somatostatin

SSTR2

somatostatin receptor 2

VEGF

vascular endothelial growth factor

References

1 

Birzniece V and Ho KKY: Mechanisms in endocrinology: Paracrine and endocrine control of the growth hormone axis by estrogen. Eur J Endocrinol. 184:R269–R278. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Caputo M, Pigni S, Agosti E, Daffara T, Ferrero A, Filigheddu N and Prodam F: Regulation of GH and GH signaling by nutrients. Cells. 10:13762021. View Article : Google Scholar : PubMed/NCBI

3 

Donato J Jr, Wasinski F, Furigo IC, Metzger M and Frazão R: Central regulation of metabolism by growth hormone. Cells. 10:1292021. View Article : Google Scholar : PubMed/NCBI

4 

Huang Z, Huang L, Waters MJ and Chen C: Insulin and growth hormone balance: Implications for obesity. Trends Endocrinol Metab. 31:642–654. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Roelfsema F, Yang RJ, Bowers CY and Veldhuis JD: Modulating effects of progesterone on spontaneous nocturnal and ghrelin-induced GH secretion in postmenopausal women. J Clin Endocrinol Metab. 104:2385–2394. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Cuny T, Graillon T, Defilles C, Datta R, Zhang S, Figarella-Branger D, Dufour H, Mougel G and Brue T: Characterization of the ability of a, second-generation SST-DA chimeric molecule, TBR-065, to suppress GH secretion from human GH-secreting adenoma cells. Pituitary. 24:351–358. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Boguszewski MCS, Carlsson M, Lindberg A, Dahlgren J, Aydin F, Camacho-Hübner C and Hokken-Koelega ACS: Near-adult height after growth hormone treatment in children born prematurely-data from KIGS. J Clin Endocrinol Metab. 105:dgaa2032020. View Article : Google Scholar : PubMed/NCBI

8 

Hjelholt AJ, Charidemou E, Griffin JL, Pedersen SB, Gudiksen A, Pilegaard H, Jessen N, Møller N and Jørgensen JOL: Insulin resistance induced by growth hormone is linked to lipolysis and associated with suppressed pyruvate dehydrogenase activity in skeletal muscle: A 2x2 factorial, randomised, crossover study in human individuals. Diabetologia. 63:2641–2653. 2020. View Article : Google Scholar : PubMed/NCBI

9 

Binder G: Short stature due to SHOX deficiency: Genotype, phenotype, and therapy. Horm Res Paediatr. 75:81–89. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Donato B and Ferreira MJ: Cardiovascular risk in turner syndrome. Rev Port Cardiol (Engl Ed). 37:607–621. 2018.In English, Portuguese. View Article : Google Scholar

11 

Muscogiuri G, Formoso G, Pugliese G, Ruggeri RM, Scarano E and Colao A: RESTARE: Prader-Willi syndrome: An uptodate on endocrine and metabolic complications. Rev Endocr Metab Disord. 20:239–250. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Roberts AE, Allanson JE, Tartaglia M and Gelb BD: Noonan syndrome. Lancet. 381:333–342. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Verkauskiene R, Petraitiene I and Albertsson Wikland K: Puberty in children born small for gestational age. Horm Res Paediatr. 80:69–77. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Guevara-Aguirre J, Guevara A, Palacios I, Pérez M, Prócel P and Terán E: GH and GHR signaling in human disease. Growth Horm IGF Res. 38:34–38. 2018. View Article : Google Scholar : PubMed/NCBI

15 

Boguszewski CL: Individual sensitivity to growth hormone replacement in adults. Rev Endocr Metab Disord. 22:117–14. 2021. View Article : Google Scholar

16 

Gasco V, Caputo M, Lanfranco F, Ghigo E and Grottoli S: Management of GH treatment in adult GH deficiency. Best Pract Res Clin Endocrinol Metab. 31:13–24. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Tritos NA and Klibanski A: Effects of growth hormone on bone. Prog Mol Biol Transl Sci. 138:193–211. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Beckers A, Petrossians P, Hanson J and Daly AF: The causes and consequences of pituitary gigantism. Nat Rev Endocrinol. 14:705–720. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Domene HM and Fierro-Carrión G: Genetic disorders of GH action pathway. Growth Horm IGF Res. 38:19–23. 2018. View Article : Google Scholar

20 

Villares R, Criado G, Juarranz Y, Lopez-Santalla M, Garcia-Cuesta EM, Rodriguez-Frade JM, Leceta J, Lucas P, Pablos JL, Martínez-A C, et al: Inhibitory role of growth hormone in the induction and progression phases of collagen-induced arthritis. Front Immunol. 9:11652018. View Article : Google Scholar : PubMed/NCBI

21 

Kopchick JJ, Berryman DE, Puri V, Lee KY and Jorgensen JOL: The effects of growth hormone on adipose tissue: Old observations, new mechanisms. Nat Rev Endocrinol. 16:135–146. 2020. View Article : Google Scholar :

22 

Shukur HH, Hussain-Alkhateeb L, Farholt S, Nørregaard O, Jørgensen AP and Hoybye C: Effects of growth hormone treatment on sleep-related parameters in adults with Prader-Willi syndrome. J Clin Endocrinol Metab. 106:e3634–e3643. 2021. View Article : Google Scholar : PubMed/NCBI

23 

Devesa J and Caicedo D: The role of growth hormone on ovarian functioning and ovarian angiogenesis. Front Endocrinol (Lausanne). 10:4502019. View Article : Google Scholar

24 

Gong Y, Luo S, Fan P, Zhu H, Li Y and Huang W: Growth hormone activates PI3K/Akt signaling and inhibits ROS accumulation and apoptosis in granulosa cells of patients with polycystic ovary syndrome. Reprod Biol Endocrinol. 18:1212020. View Article : Google Scholar : PubMed/NCBI

25 

Tesarik J, Galán-Lázaro M, Conde-López C, Chiara-Rapisarda AM and Mendoza-Tesarik R: The effect of GH administration on oocyte and zygote quality in young women with repeated implantation failure after IVF. Front Endocrinol (Lausanne). 11:5195722020. View Article : Google Scholar

26 

Subramani R, Nandy SB, Pedroza DA and Lakshmanaswamy R: Role of growth hormone in breast cancer. Endocrinology. 158:1543–1555. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Coker-Gurkan A, Celik M, Ugur M, Arisan ED, Obakan-Yerlikaya P, Durdu ZB and Palavan-Unsal N: Curcumin inhibits autocrine growth hormone-mediated invasion and metastasis by targeting NF-κB signaling and polyamine metabolism in breast cancer cells. Amino Acids. 50:1045–1069. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Zhu X, Li Y, Xu G and Fu C: Growth hormone receptor promotes breast cancer progression via the BRAF/MEK/ERK signaling pathway. FEBS Open Bio. 10:1013–1020. 2020. View Article : Google Scholar : PubMed/NCBI

29 

Zhang N, Ng AS, Cai S, Li Q, Yang L and Kerr D: Novel therapeutic strategies: Targeting epithelial-mesenchymal transition in colorectal cancer. Lancet Oncol. 22:e358–e368. 2021. View Article : Google Scholar : PubMed/NCBI

30 

Lambert AW and Weinberg RA: Linking EMT programmes to normal and neoplastic epithelial stem cells. Nat Rev Cancer. 21:325–338. 2021. View Article : Google Scholar : PubMed/NCBI

31 

Yang J, Antin P, Berx G, Blanpain C, Brabletz T, Bronner M, Campbell K, Cano A, Casanova J, Christofori G, et al: Guidelines and definitions for research on epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 21:341–352. 2020. View Article : Google Scholar : PubMed/NCBI

32 

Baskari S, Govatati S, Madhuri V, Nallabelli N, K PM, Naik S, Poornachandar, Balka S, Tamanam RR and Devi VR: Influence of autocrine growth hormone on NF-κB activation leading to epithelial-mesenchymal transition of mammary carcinoma. Tumour Biol. 39:10104283177191212017. View Article : Google Scholar

33 

Chesnokova V and Melmed S: Growth hormone in the tumor microenvironment. Arch Endocrinol Metab. 63:568–575. 2019. View Article : Google Scholar

34 

Brittain AL, Basu R, Qian Y and Kopchick JJ: Growth hormone and the epithelial-to-mesenchymal transition. J Clin Endocrinol Metab. 102:3662–3673. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Chen YJ, You ML, Chong QY, Pandey V, Zhuang QS, Liu DX, Ma L, Zhu T and Lobie PE: Autocrine human growth hormone promotes invasive and cancer stem cell-like behavior of hepatocellular carcinoma cells by STAT3 dependent inhibition of CLAUDIN-1 expression. Int J Mol Sci. 18:12742017. View Article : Google Scholar :

36 

Neggers SJ, Muhammad A and van der Lely AJ: Pegvisomant treatment in acromegaly. Neuroendocrinology. 103:59–65. 2016. View Article : Google Scholar

37 

Tritos NA and Biller BM: Pegvisomant: A growth hormone receptor antagonist used in the treatment of acromegaly. Pituitary. 20:129–135. 2017. View Article : Google Scholar

38 

Kuhn E and Chanson P: Cabergoline in acromegaly. Pituitary. 20:121–128. 2017. View Article : Google Scholar

39 

Chanson P: Medical treatment of acromegaly with dopamine agonists or somatostatin analogs. Neuroendocrinology. 103:50–58. 2016. View Article : Google Scholar

40 

Maffezzoni F, Formenti AM, Mazziotti G, Frara S and Giustina A: Current and future medical treatments for patients with acromegaly. Expert Opin Pharmacother. 17:1631–1642. 2016. View Article : Google Scholar : PubMed/NCBI

41 

Wang JW, Li Y, Mao ZG, Hu B, Jiang XB, Song BB, Wang X, Zhu YH and Wang HJ: Clinical applications of somatostatin analogs for growth hormone-secreting pituitary adenomas. Patient Prefer Adherence. 8:43–51. 2014.PubMed/NCBI

42 

Colao A, Grasso LFS, Giustina A, Melmed S, Chanson P, Pereira AM and Pivonello R: Acromegaly. Nat Rev Dis Primers. 5:202019. View Article : Google Scholar : PubMed/NCBI

43 

Valea A, Ghervan C, Carsote M, Morar A, Iacob I, Tomesc F, Pop DD and Georgescu C: Effects of combination therapy: Somatostatin analogues and dopamine agonists on GH and IGF1 levels in acromegaly. Clujul Med. 88:310–313. 2015.

44 

Augustine RA, Ladyman SR, Bouwer GT, Alyousif Y, Sapsford TJ, Scott V, Kokay IC, Grattan DR and Brown CH: Prolactin regulation of oxytocin neurone activity in pregnancy and lactation. J Physiol. 595:3591–3605. 2017. View Article : Google Scholar : PubMed/NCBI

45 

Chen Z, Luo J, Zhang C, Ma Y, Sun S, Zhang T and Loor JJ: Mechanism of prolactin inhibition of miR-135b via methylation in goat mammary epithelial cells. J Cell Physiol. 233:651–662. 2018. View Article : Google Scholar

46 

Borba VV, Zandman-Goddard G and Shoenfeld Y: Prolactin and autoimmunity. Front Immunol. 9:732018. View Article : Google Scholar : PubMed/NCBI

47 

García-Rizo C, Vázquez-Bourgon J, Labad J, Ortiz García de la Foz V, Gómez-Revuelta M, Juncal Ruiz M and Crespo-Facorro B: Prolactin, metabolic and immune parameters in naïve subjects with a first episode of psychosis. Prog Neuropsychopharmacol Biol Psychiatry. 110:1103322021. View Article : Google Scholar

48 

Bernard V, Young J and Binart N: Prolactin-a pleiotropic factor in health and disease. Nat Rev Endocrinol. 15:356–365. 2019. View Article : Google Scholar

49 

Bernard V, Young J, Chanson P and Binart N: New insights in prolactin: Pathological implications. Nat Rev Endocrinol. 11:265–275. 2015. View Article : Google Scholar

50 

Moghbeli M: Genetics of recurrent pregnancy loss among Iranian population. Mol Genet Genomic Med. 7:e8912019. View Article : Google Scholar :

51 

Kavarthapu R and Dufau ML: Essential role of endogenous prolactin and CDK7 in estrogen-induced upregulation of the prolactin receptor in breast cancer cells. Oncotarget. 8:27353–27363. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Ezoe K, Miki T, Ohata K, Fujiwara N, Yabuuchi A, Kobayashi T and Kato K: Prolactin receptor expression and its role in trophoblast outgrowth in human embryos. Reprod Biomed Online. 42:699–707. 2021. View Article : Google Scholar

53 

Mestre Citrinovitz AC, Langer L, Strowitzki T and Germeyer A: Resveratrol enhances decidualization of human endometrial stromal cells. Reproduction. 159:453–463. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Napso T, Yong HEJ, Lopez-Tello J and Sferruzzi-Perri AN: The role of placental hormones in mediating maternal adaptations to support pregnancy and lactation. Front Physiol. 9:10912018. View Article : Google Scholar : PubMed/NCBI

55 

Kalu E, Bhaskaran S, Thum MY, Vishwanatha R, Croucher C, Sherriff E, Ford B and Bansal AS: Serial estimation of Th1:th2 cytokines profile in women undergoing in-vitro fertilization-embryo transfer. Am J Reprod Immunol. 59:206–211. 2008. View Article : Google Scholar : PubMed/NCBI

56 

Soh MC and Moretto M: The use of biologics for autoimmune rheumatic diseases in fertility and pregnancy. Obstet Med. 13:5–13. 2020. View Article : Google Scholar : PubMed/NCBI

57 

Borba VV, Zandman-Goddard G and Shoenfeld Y: Prolactin and autoimmunity: The hormone as an inflammatory cytokine. Best Pract Res Clin Endocrinol Metab. 33:1013242019. View Article : Google Scholar

58 

Proietto S, Cortasa SA, Corso MC, Inserra PIF, Charif SE, Schmidt AR, Di Giorgio NP, Lux-Lantos V, Vitullo AD, Dorfman VB and Halperin J: Prolactin is a strong candidate for the regulation of luteal steroidogenesis in vizcachas (Lagostomus maximus). Int J Endocrinol. 2018:19106722018. View Article : Google Scholar :

59 

Trott JF, Schennink A, Petrie WK, Manjarin R, VanKlompenberg MK and Hovey RC: Triennial lactation symposium: Prolactin: The multifaceted potentiator of mammary growth and function. J Anim Sci. 90:1674–1686. 2012. View Article : Google Scholar

60 

Chen Y, Moutal A, Navratilova E, Kopruszinski C, Yue X, Ikegami M, Chow M, Kanazawa I, Bellampalli SS, Xie J, et al: The prolactin receptor long isoform regulates nociceptor sensitization and opioid-induced hyperalgesia selectively in females. Sci Transl Med. 12:eaay75502020. View Article : Google Scholar : PubMed/NCBI

61 

Anderson MG, Zhang Q, Rodriguez LE, Hecquet CM, Donawho CK, Ansell PJ, Ansell PJ and Reilly EB: ABBV-176, a PRLR antibody drug conjugate with a potent DNA-damaging PBD cytotoxin and enhanced activity with PARP inhibition. BMC Cancer. 21:6812021. View Article : Google Scholar

62 

Li D, San M, Zhang J, Yang A, Xie W, Chen Y, Lu X, Zhang Y, Zhao M, Feng X and Zheng Y: Oxytocin receptor induces mammary tumorigenesis through prolactin/p-STAT5 pathway. Cell Death Dis. 12:5882021. View Article : Google Scholar : PubMed/NCBI

63 

Borcherding DC, Hugo ER, Fox SR, Jacobson EM, Hunt BG, Merino EJ and Ben-Jonathan N: Suppression of breast cancer by small molecules that block the prolactin receptor. Cancers (Basel). 13:26622021. View Article : Google Scholar

64 

O'Leary KA, Rugowski DE, Shea MP, Sullivan R, Moser AR and Schuler LA: Prolactin synergizes with canonical Wnt signals to drive development of ER+ mammary tumors via activation of the Notch pathway. Cancer Lett. 503:231–239. 2021. View Article : Google Scholar : PubMed/NCBI

65 

Campbell KM, O'Leary KA, Rugowski DE, Mulligan WA, Barnell EK, Skidmore ZL, Krysiak K, Griffith M, Schuler LA and Griffith OL: A spontaneous aggressive ERα+ mammary tumor model is driven by Kras activation. Cell Rep. 28:1526–1537.e4. 2019. View Article : Google Scholar

66 

MacDonald TM, Thomas LN, Daze E, Marignani P, Barnes PJ and Too CK: Prolactin-inducible EDD E3 ubiquitin ligase promotes TORC1 signalling, anti-apoptotic protein expression, and drug resistance in breast cancer cells. Am J Cancer Res. 9:1484–1503. 2019.PubMed/NCBI

67 

Chen X, Wu D, Zheng Y, Liu X and Wang J: Preparation of a growth hormone receptor/prolactin receptor bispecific antibody antagonist which exhibited anti-cancer activity. Front Pharmacol. 11:5984232020. View Article : Google Scholar : PubMed/NCBI

68 

Dandawate P, Kaushik G, Ghosh C, Standing D, Ali Sayed AA, Choudhury S, Subramaniam D, Manzardo A, Banerjee T, Santra S, et al: Diphenylbutylpiperidine antipsychotic drugs inhibit prolactin receptor signaling to reduce growth of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 158:1433–1449.e27. 2020. View Article : Google Scholar

69 

Ramirez-Hernandez G, Adan-Castro E, Diaz-Lezama N, Ruiz-Herrera X, Martinez de la Escalera G, Macotela Y and Clapp C: Global deletion of the prolactin receptor aggravates streptozotocin-induced diabetes in mice. Front Endocrinol (Lausanne). 12:6196962021. View Article : Google Scholar

70 

Wen Y, Wang Y, Chelariu-Raicu A, Stur E, Liu Y, Corvigno S, Bartsch F, Redfern L, Zand B, Kang Y, et al: Blockade of the short form of prolactin receptor induces FOXO3a/EIF-4EBP1-mediated cell death in uterine cancer. Mol Cancer Ther. 19:1943–1954. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Asad AS, Nicola Candia AJ, Gonzalez N, Zuccato CF, Seilicovich A and Candolfi M: The role of the prolactin receptor pathway in the pathogenesis of glioblastoma: What do we know so far? Expert Opin Ther Targets. 24:1121–1133. 2020. View Article : Google Scholar

72 

Boutillon F, Pigat N, Sala LS, Reyes-Gomez E, Moriggl R, Guidotti JE and Goffin V: STAT5a/b deficiency delays, but does not prevent, prolactin-driven prostate tumorigenesis in mice. Cancers (Basel). 11:9292019. View Article : Google Scholar

73 

ivero-Segura NA, Flores-Soto E, García de la Cadena S, Coronado-Mares I, Gomez-Verjan JC, Ferreira DG, Cabrera-Reyes EA, Lopes LV, Massieu L and Cerbón M: Prolactin-induced neuroprotection against glutamate excitotoxicity is mediated by the reduction of [Ca2+]i overload and NF-κB activation. PLoS One. 12:e01769102017. View Article : Google Scholar

74 

Yousefvand S, Hadjzadeh MA, Vafaee F and Dolatshad H: The protective effects of prolactin on brain injury. Life Sci. 263:1185472020. View Article : Google Scholar : PubMed/NCBI

75 

Lopez-Vicchi F, De Winne C, Brie B, Sorianello E, Ladyman SR and Becu-Villalobos D: Metabolic functions of prolactin: Physiological and pathological aspects. J Neuroendocrinol. 32:e128882020. View Article : Google Scholar

76 

Charoenphandhu N and Krishnamra N: Prolactin is an important regulator of intestinal calcium transport. Can J Physiol Pharmacol. 85:569–581. 2007. View Article : Google Scholar : PubMed/NCBI

77 

Ponce AJ, Galván-Salas T, Lerma-Alvarado RM, Ruiz-Herrera X, Hernández-Cortés T, Valencia-Jiménez R, Cárdenas-Rodríguez LE, Martínez de la Escalera G, Clapp C and Macotela Y: Low prolactin levels are associated with visceral adipocyte hypertrophy and insulin resistance in humans. Endocrine. 67:331–343. 2020. View Article : Google Scholar : PubMed/NCBI

78 

Tatum RC, McGowan CM and Ireland JL: Efficacy of pergolide for the management of equine pituitary pars intermedia dysfunction: A systematic review. Vet J. 266:1055622020. View Article : Google Scholar : PubMed/NCBI

79 

Harris K, Murphy KE, Horn D, MacGilivray J and Yudin MH: Safety of cabergoline for postpartum lactation inhibition or suppression: A systematic review. J Obstet Gynaecol Can. 42:308–315.e20. 2020. View Article : Google Scholar

80 

Krysiak R and Okopień B: Sexual functioning in hyperprolactinemic patients treated with cabergoline or bromocriptine. Am J Ther. 26:e433–e440. 2019. View Article : Google Scholar

81 

Khalil G, Khan FA, Jamal QM, Saleem A, Masroor H and Abbas K: Change in insulin sensitivity and lipid profile after dopamine agonist therapy in patients with prolactinoma. Cureus. 13:e178242021.PubMed/NCBI

82 

Peuskens J, Pani L, Detraux J and De Hert M: The effects of novel and newly approved antipsychotics on serum prolactin levels: A comprehensive review. CNS Drugs. 28:421–453. 2014.PubMed/NCBI

83 

Drobnis EZ and Nangia AK: Psychotropics and male reproduction. Adv Exp Med Biol. 1034:63–101. 2017. View Article : Google Scholar : PubMed/NCBI

84 

Safer DJ, Calarge CA and Safer AM: Prolactin serum concentrations during aripiprazole treatment in youth. J Child Adolesc Psychopharmacol. 23:282–289. 2013. View Article : Google Scholar : PubMed/NCBI

85 

Davis JR and McNeilly AS: Is pituitary gene therapy realistic? Clin Endocrinol (Oxf). 55:427–433. 2001. View Article : Google Scholar

86 

Filatov M, Khramova Y, Parshina E, Bagaeva T and Semenova M: Influence of gonadotropins on ovarian follicle growth and development in vivo and in vitro. Zygote. 25:235–243. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Ulloa-Aguirre A and Lira-Albarran S: Clinical applications of gonadotropins in the male. Prog Mol Biol Transl Sci. 143:121–174. 2016. View Article : Google Scholar : PubMed/NCBI

88 

Das N and Kumar TR: Molecular regulation of follicle-stimulating hormone synthesis, secretion and action. J Mol Endocrinol. 60:R131–R155. 2018. View Article : Google Scholar : PubMed/NCBI

89 

Casarini L, Santi D, Brigante G and Simoni M: Two hormones for one receptor: Evolution, biochemistry, actions, and pathophysiology of LH and hCG. Endocr Rev. 39:549–592. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Troppmann B, Kleinau G, Krause G and Gromoll J: Structural and functional plasticity of the luteinizing hormone/choriogonadotrophin receptor. Hum Reprod Update. 19:583–602. 2013. View Article : Google Scholar : PubMed/NCBI

91 

Son WY, Das M, Shalom-Paz E and Holzer H: Mechanisms of follicle selection and development. Minerva Ginecol. 63:89–102. 2011.PubMed/NCBI

92 

Themmen APN and Huhtaniemi IT: Mutations of gonadotropins and gonadotropin receptors: Elucidating the physiology and pathophysiology of pituitary-gonadal function. Endocr Rev. 21:551–583. 2000. View Article : Google Scholar : PubMed/NCBI

93 

Duan J, Xu P, Cheng X, Mao C, Croll T, He X, Shi J, Luan X, Yin W, You E, et al: Structures of full-length glycoprotein hormone receptor signalling complexes. Nature. 598:688–692. 2021. View Article : Google Scholar : PubMed/NCBI

94 

Jiang X, Dias JA and He X: Structural biology of glycoprotein hormones and their receptors: Insights to signaling. Mol Cell Endocrinol. 382:424–451. 2014. View Article : Google Scholar

95 

Abbara A, Clarke SA and Dhillo WS: Clinical potential of kisspeptin in reproductive health. Trends Mol Med. 27:807–823. 2021. View Article : Google Scholar : PubMed/NCBI

96 

Skorupskaite K and Anderson RA: Hypothalamic neurokinin signalling and its application in reproductive medicine. Pharmacol Ther. 230:1079602022. View Article : Google Scholar

97 

Messina A, Pulli K, Santini S, Acierno J, Känsäkoski J, Cassatella D, Xu C, Casoni F, Malone SA, Ternier G, et al: Neuron-derived neurotrophic factor is mutated in congenital hypogonadotropic hypogonadism. Am J Hum Genet. 106:58–70. 2020. View Article : Google Scholar :

98 

Vanacker C, Defazio RA, Sykes CM and Moenter SM: A role for glial fibrillary acidic protein (GFAP)-expressing cells in the regulation of gonadotropin-releasing hormone (GnRH) but not arcuate kisspeptin neuron output in male mice. Elife. 10:e682052021. View Article : Google Scholar : PubMed/NCBI

99 

Uenoyama Y, Nagae M, Tsuchida H, Inoue N and Tsukamura H: Role of KNDy neurons expressing kisspeptin, neurokinin B, and dynorphin A as a GnRH pulse generator controlling mammalian reproduction. Front Endocrinol (Lausanne). 12:7246322021. View Article : Google Scholar

100 

Hughes CHK and Murphy BD: Nuclear receptors: Key regulators of somatic cell functions in the ovulatory process. Mol Aspects Med. 78:1009372021. View Article : Google Scholar

101 

Abreu AP, Toro CA, Song YB, Navarro VM, Bosch MA, Eren A, Liang JN, Carroll RS, Latronico AC, Rønnekleiv OK, et al: MKRN3 inhibits the reproductive axis through actions in kisspeptin-expressing neurons. J Clin Invest. 130:4486–4500. 2020.PubMed/NCBI

102 

Li M, Chen Y, Liao B, Tang J, Zhong J and Lan D: The role of kisspeptin and MKRN3 in the diagnosis of central precocious puberty in girls. Endocr Connect. 10:1147–1154. 2021. View Article : Google Scholar : PubMed/NCBI

103 

Sertorio MN, Estadella D, Ribeiro DA and Pisani LP: Could parental high-fat intake program the reproductive health of male offspring? A review. Crit Rev Food Sci Nutr. 1–8. 2021.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

104 

Santoro N, Schauer IE, Kuhn K, Fought AJ, Babcock-Gilbert S and Bradford AP: Gonadotropin response to insulin and lipid infusion reproduces the reprometabolic syndrome of obesity in eumenorrheic lean women: A randomized crossover trial. Fertil Steril. 116:566–574. 2021. View Article : Google Scholar : PubMed/NCBI

105 

Hsueh AJ and He J: Gonadotropins and their receptors: Coevolution, genetic variants, receptor imaging, and functional antagonists. Biol Reprod. 99:3–12. 2018. View Article : Google Scholar : PubMed/NCBI

106 

Chu YL, Xu YR, Yang WX and Sun Y: The role of FSH and TGF-β superfamily in follicle atresia. Aging (Albany NY). 10:305–321. 2018. View Article : Google Scholar

107 

Smitz J, Wolfenson C, Chappel S and Ruman J: Follicle-stimulating hormone: A review of form and function in the treatment of infertility. Reprod Sci. 23:706–716. 2016. View Article : Google Scholar

108 

di Clemente N, Racine C, Pierre A and Taieb J: Anti-Müllerian hormone in female reproduction. Endocr Rev. 42:753–782. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Mills EG, Yang L, Nielsen MF, Kassem M, Dhillo WS and Comninos AN: The relationship between bone and reproductive hormones beyond estrogens and androgens. Endocr Rev. 42:691–719. 2021. View Article : Google Scholar : PubMed/NCBI

110 

Zhu D, Li X, Macrae VE, Simoncini T and Fu X: Extragonadal effects of follicle-stimulating hormone on osteoporosis and cardiovascular disease in women during menopausal transition. Trends Endocrinol Metab. 29:571–580. 2018. View Article : Google Scholar : PubMed/NCBI

111 

Chin KY: The relationship between follicle-stimulating hormone and bone health: Alternative explanation for bone loss beyond oestrogen? Int J Med Sci. 15:1373–1383. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Liu WX, Zhang YJ, Wang YF, Klinger FG, Tan SJ, Farini D, De Felici M, Shen W and Cheng SF: Protective mechanism of luteinizing hormone and follicle-stimulating hormone against nicotine-induced damage of mouse early folliculogenesis. Front Cell Dev Biol. 9:7233882021. View Article : Google Scholar : PubMed/NCBI

113 

Kumariya S, Ubba V, Jha RK and Gayen JR: Autophagy in ovary and polycystic ovary syndrome: Role, dispute and future perspective. Autophagy. 17:2706–2733. 2021. View Article : Google Scholar : PubMed/NCBI

114 

Guo Y, Zhao M, Bo T, Ma S, Yuan Z, Chen W, He Z, Hou X, Liu J, Zhang Z, et al: Blocking FSH inhibits hepatic cholesterol biosynthesis and reduces serum cholesterol. Cell Res. 29:151–166. 2019. View Article : Google Scholar :

115 

Veldhuis-Vlug AG, Woods GN, Sigurdsson S, Ewing SK, Le PT, Hue TF, Vittinghoff E, Xu K, Gudnason V, Sigurdsson G, et al: Serum FSH is associated with BMD, bone marrow adiposity, and body composition in the AGES-Reykjavik study of older adults. J Clin Endocrinol Metab. 106:e1156–e1169. 2021. View Article : Google Scholar :

116 

Wu KC, Ewing SK, Li X, Sigurðsson S, Guðnason V, Kado DM, Hue TF, Woods GN, Veldhuis-Vlug AG, Vittinghoff E, et al: FSH level and changes in bone mass and body composition in older women and men. J Clin Endocrinol Metab. 106:2876–2889. 2021. View Article : Google Scholar : PubMed/NCBI

117 

Bloise E, Ciarmela P, Dela Cruz C, Luisi S, Petraglia F and Reis FM: Activin A in mammalian physiology. Physiol Rev. 99:739–780. 2019. View Article : Google Scholar

118 

Bernard DJ, Smith CL and Brûlé E: A tale of two proteins: Betaglycan, IGSF1, and the continuing search for the inhibin B receptor. Trends Endocrinol Metab. 31:37–45. 2020. View Article : Google Scholar

119 

Peng YJ, Yu H, Hao X, Dong W, Yin X, Lin M, Zheng J and Zhou BO: Luteinizing hormone signaling restricts hematopoietic stem cell expansion during puberty. EMBO J. 37:e989842018. View Article : Google Scholar : PubMed/NCBI

120 

Del Castillo LM, Buigues A, Rossi V, Soriano MJ, Martinez J, De Felici M, Lamsira HK, Di Rella F, Klinger FG, Pellicer A and Herraiz S: The cyto-protective effects of LH on ovarian reserve and female fertility during exposure to gonadotoxic alkylating agents in an adult mouse model. Hum Reprod. 36:2514–2528. 2021. View Article : Google Scholar : PubMed/NCBI

121 

Xi G, An L, Wang W, Hao J, Yang Q, Ma L, Lu J, Wang Y, Wang W, Zhao W, et al: The mRNA-destabilizing protein tristetraprolin targets 'meiosis arrester' Nppc mRNA in mammalian preovulatory follicles. Proc Natl Acad Sci USA. 118:e20183451182021. View Article : Google Scholar

122 

Dalle IA, Paranal R, Zarka J, Paul S, Sasaki K, Li W, Ning J, Short NJ, Ohanian M, Cortes JE, et al: Impact of luteinizing hormone suppression on hematopoietic recovery after intensive chemotherapy in patients with leukemia. Haematologica. 106:1097–1105. 2021.

123 

Elias HK and Van den Brink MRM: New option for improving hematological recovery: Suppression of luteinizing hormone. Haematologica. 106:929–931. 2021.

124 

Navarro VM: Metabolic regulation of kisspeptin-the link between energy balance and reproduction. Nat Rev Endocrinol. 16:407–420. 2020. View Article : Google Scholar : PubMed/NCBI

125 

Duffy DM, Ko C, Jo M, Brannstrom M and Curry TE: Ovulation: Parallels with inflammatory processes. Endocr Rev. 40:369–416. 2019. View Article : Google Scholar :

126 

Rossi V, Lispi M, Longobardi S, Mattei M, Di Rella F, Salustri A, De Felici M and Klinger FG: LH prevents cisplatin-induced apoptosis in oocytes and preserves female fertility in mouse. Cell Death Differ. 24:72–82. 2017. View Article : Google Scholar :

127 

Li X, Zhou L, Peng G, Liao M, Zhang L, Hu H, Long L, Tang X, Qu H, Shao J, et al: Pituitary P62 deficiency leads to female infertility by impairing luteinizing hormone production. Exp Mol Med. 53:1238–1249. 2021. View Article : Google Scholar : PubMed/NCBI

128 

Blair JA, Bhatta S, McGee H and Casadesus G: Luteinizing hormone: Evidence for direct action in the CNS. Horm Behav. 76:57–62. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Burnham VL and Thornton JE: Luteinizing hormone as a key player in the cognitive decline of Alzheimer's disease. Horm Behav. 76:48–56. 2015. View Article : Google Scholar : PubMed/NCBI

130 

Natanzon Y, Goode EL and Cunningham JM: Epigenetics in ovarian cancer. Semin Cancer Biol. 51:160–169. 2018. View Article : Google Scholar :

131 

Kossaï M, Leary A, Scoazec JY and Genestie C: Ovarian cancer: A heterogeneous disease. Pathobiology. 85:41–49. 2018. View Article : Google Scholar

132 

Cheung J, Lokman NA, Abraham RD, Macpherson AM, Lee E, Grutzner F, Ghinea N, Oehler MK and Ricciardelli C: Reduced gonadotrophin receptor expression is associated with a more aggressive ovarian cancer phenotype. Int J Mol Sci. 22:712020. View Article : Google Scholar

133 

Wang Z and Dong C: Gluconeogenesis in cancer: Function and regulation of PEPCK, FBPase, and G6Pase. Trends Cancer. 5:30–45. 2019. View Article : Google Scholar : PubMed/NCBI

134 

MacLean DM and Jayaraman V: Acid-sensing ion channels are tuned to follow high-frequency stimuli. J Physiol. 594:2629–2645. 2016. View Article : Google Scholar : PubMed/NCBI

135 

Wu J, Leng T, Jing L, Jiang N, Chen D, Hu Y, Xiong ZG and Zha XM: Two di-leucine motifs regulate trafficking and function of mouse ASIC2a. Mol Brain. 9:92016. View Article : Google Scholar : PubMed/NCBI

136 

Dong HW, Wang K, Chang XX, Jin FF, Wang Q, Jiang XF, Liu JR, Wu YH and Yang C: Beta-ionone-inhibited proliferation of breast cancer cells by inhibited COX-2 activity. Arch Toxicol. 93:2993–3003. 2019. View Article : Google Scholar : PubMed/NCBI

137 

Echizen K, Hirose O, Maeda Y and Oshima M: Inflammation in gastric cancer: Interplay of the COX-2/prostaglandin E2 and Toll-like receptor/MyD88 pathways. Cancer Sci. 107:391–397. 2016. View Article : Google Scholar : PubMed/NCBI

138 

Davenport JR, Cai Q, Ness RM, Milne G, Zhao Z, Smalley WE, Zheng W and Shrubsole MJ: Evaluation of pro-inflammatory markers plasma C-reactive protein and urinary prostaglandin-E2 metabolite in colorectal adenoma risk. Mol Carcinog. 55:1251–1261. 2016. View Article : Google Scholar :

139 

Feng D, Zhao T, Yan K, Liang H, Liang J, Zhou Y, Zhao W and Ling B: Gonadotropins promote human ovarian cancer cell migration and invasion via a cyclooxygenase 2-dependent pathway. Oncol Rep. 38:1091–1098. 2017. View Article : Google Scholar : PubMed/NCBI

140 

Lau MT, Wong AS and Leung PC: Gonadotropins induce tumor cell migration and invasion by increasing cyclooxygenases expression and prostaglandin E(2) production in human ovarian cancer cells. Endocrinology. 151:2985–2993. 2010. View Article : Google Scholar : PubMed/NCBI

141 

Li S, Ji X, Wang R and Miao Y: Follicle-stimulating hormone promoted pyruvate kinase isozyme type M2-induced glycolysis and proliferation of ovarian cancer cells. Arch Gynecol Obstet. 299:1443–1451. 2019. View Article : Google Scholar : PubMed/NCBI

142 

Zhang J, Zhang J, Wei Y, Li Q and Wang Q: ACTL6A regulates follicle-stimulating hormone-driven glycolysis in ovarian cancer cells via PGK1. Cell Death Dis. 10:8112019. View Article : Google Scholar : PubMed/NCBI

143 

Perez-Juarez CE, Arechavaleta-Velasco F, Mendez C and Díaz-Cueto L: Progranulin expression induced by follicle-stimulating hormone in ovarian cancer cell lines depends on the histological subtype. Med Oncol. 37:592020. View Article : Google Scholar : PubMed/NCBI

144 

Chen J, Bai M, Ning C, Xie B, Zhang J, Liao H, Xiong J, Tao X, Yan D, Xi X, et al: Gankyrin facilitates follicle-stimulating hormone-driven ovarian cancer cell proliferation through the PI3K/AKT/HIF-1α/cyclin D1 pathway. Oncogene. 35:2506–2517. 2016. View Article : Google Scholar

145 

Zhang M, Zhang M, Wang J, Cai Q, Zhao R, Yu Y, Tai H, Zhang X and Xu C: Retro-inverso follicle-stimulating hormone peptide-mediated polyethylenimine complexes for targeted ovarian cancer gene therapy. Drug Deliv. 25:995–1003. 2018. View Article : Google Scholar : PubMed/NCBI

146 

Zhang MX, Hong SS, Cai QQ, Zhang M, Chen J, Zhang XY and Xu CJ: Transcriptional control of the MUC16 promoter facilitates follicle-stimulating hormone peptide-conjugated shRNA nanoparticle-mediated inhibition of ovarian carcinoma in vivo. Drug Deliv. 25:797–806. 2018. View Article : Google Scholar : PubMed/NCBI

147 

Jiang N, Wu J, Leng T, Yang T, Zhou Y, Jiang Q, Wang B, Hu Y, Ji YH, Simon RP, et al: Region specific contribution of ASIC2 to acidosis-and ischemia-induced neuronal injury. J Cereb Blood Flow Metab. 37:528–540. 2017. View Article : Google Scholar :

148 

Liao H, Zhou Q, Gu Y, Duan T and Feng Y: Luteinizing hormone facilitates angiogenesis in ovarian epithelial tumor cells and metformin inhibits the effect through the mTOR signaling pathway. Oncol Rep. 27:1873–1888. 2012.PubMed/NCBI

149 

Garrido MP, Bruneau N, Vega M, Selman A, Tapia JC and Romero C: Follicle-stimulating hormone promotes nerve growth factor and vascular endothelial growth factor expression in epithelial ovarian cells. Histol Histopathol. 35:961–971. 2020.PubMed/NCBI

150 

Zhang J, Sun YF, Xu YM, Shi BJ, Han Y, Luo ZY, Zhao ZM, Hao GM and Gao BL: Effect of endometrium thickness on clinical outcomes in luteal phase short-acting GnRH-a long protocol and GnRH-Ant protocol. Front Endocrinol (Lausanne). 12:5787832021. View Article : Google Scholar

151 

Sauerbrun-Cutler MT and Alvero R: Short- and long-term impact of gonadotropin-releasing hormone analogue treatment on bone loss and fracture. Fertil Steril. 112:799–803. 2019. View Article : Google Scholar : PubMed/NCBI

152 

Tepekoy F, Uysal F, Acar N, Ustunel I and Akkoyunlu G: The effect of GnRH antagonist cetrorelix on Wnt signaling members in pubertal and adult mouse ovaries. Histochem Cell Biol. 152:423–437. 2019. View Article : Google Scholar : PubMed/NCBI

153 

Doroszko M, Chrusciel M, Stelmaszewska J, Slezak T, Anisimowicz S, Plöckinger U, Quinkler M, Bonomi M, Wolczynski S, Huhtaniemi I, et al: GnRH antagonist treatment of malignant adrenocortical tumors. Endocr Relat Cancer. 26:103–117. 2019. View Article : Google Scholar

154 

Xu H, Zhao S, Gao X, Wu X, Xia L, Zhang D, Li J, Zhang A and Xu B: GnRH antagonist protocol with cessation of cetrorelix on trigger day improves embryological outcomes for patients with sufficient ovarian reserve. Front Endocrinol (Lausanne). 12:7588962021. View Article : Google Scholar

155 

Practice Committee of the American Society for Reproductive Medicine. Electronic address: asrm@asrm.org: Practice Committee of the American Society for Reproductive Medicine: Evidence-based treatments for couples with unexplained infertility: A guideline. Fertil Steril. 113:305–322. 2020. View Article : Google Scholar

156 

Krzastek SC, Sharma D, Abdullah N, Sultan M, Machen GL, Wenzel JL, Ells A, Chen X, Kavoussi M, Costabile RA, et al: Long-term safety and efficacy of clomiphene citrate for the treatment of hypogonadism. J Urol. 202:1029–1035. 2019. View Article : Google Scholar : PubMed/NCBI

157 

Miller GD, Moore C, Nair V, Hill B, Willick SE, Rogol AD and Eichner D: Hypothalamic-pituitary-testicular axis effects and urinary detection following clomiphene administration in males. J Clin Endocrinol Metab. 104:906–914. 2019. View Article : Google Scholar

158 

Kirshenbaum M, Haas J, Nahum R, Aizer A, Yinon Y and Orvieto R: The effect of ovarian stimulation on endothelial function-A prospective cohort study using peripheral artery tonometry. J Clin Endocrinol Metab. 105:dgaa6812020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

October-2022
Volume 50 Issue 4

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Tian W, Qi H, Wang Z, Qiao S, Wang P, Dong J and Wang H: Hormone supply to the pituitary gland: A comprehensive investigation of female‑related tumors (Review). Int J Mol Med 50: 122, 2022
APA
Tian, W., Qi, H., Wang, Z., Qiao, S., Wang, P., Dong, J., & Wang, H. (2022). Hormone supply to the pituitary gland: A comprehensive investigation of female‑related tumors (Review). International Journal of Molecular Medicine, 50, 122. https://doi.org/10.3892/ijmm.2022.5178
MLA
Tian, W., Qi, H., Wang, Z., Qiao, S., Wang, P., Dong, J., Wang, H."Hormone supply to the pituitary gland: A comprehensive investigation of female‑related tumors (Review)". International Journal of Molecular Medicine 50.4 (2022): 122.
Chicago
Tian, W., Qi, H., Wang, Z., Qiao, S., Wang, P., Dong, J., Wang, H."Hormone supply to the pituitary gland: A comprehensive investigation of female‑related tumors (Review)". International Journal of Molecular Medicine 50, no. 4 (2022): 122. https://doi.org/10.3892/ijmm.2022.5178