scispace - formally typeset
Search or ask a question

Showing papers on "Prolactin published in 2011"


Journal ArticleDOI
TL;DR: Understanding subcellular mechanisms that underlie pituitary tumorigenesis will enable development of tumor aggression markers as well as novel targeted therapies.
Abstract: Pituitary adenomas may hypersecrete hormones (including prolactin, growth hormone and adrenocorticotropic hormone, and rarely follicle-stimulating hormone, luteinizing hormone or TSH) or may be nonfunctional. Despite their high prevalence in the general population, these tumors are invariably benign and exhibit features of differentiated pituitary cell function as well as premature proliferative arrest. Pathogenesis of dysregulated pituitary cell proliferation and unrestrained hormone hypersecretion may be mediated by hypothalamic, intrapituitary and/or peripheral factors. Altered expression of pituitary cell cycle genes, activation of pituitary selective oncoproteins or loss of pituitary suppressor factors may be associated with aberrant growth factor signaling. Considerable information on the etiology of these tumors has been derived from transgenic animal models, which may not accurately and universally reflect human tumor pathophysiology. Understanding subcellular mechanisms that underlie pituitary tumorigenesis will enable development of tumor aggression markers as well as novel targeted therapies.

365 citations


Journal ArticleDOI
TL;DR: Endocrine sequences for dog cycles are not unlike those of many other mammals, including selection of ovulatory follicles by increased LH pulsatility, the occurrence of estrus behavior and LH surge during a decline in the estrogen: progestin ratio, a pronounced preovulatory luteinization as in humans and rodents, and luteotrophic roles for both LH and prolactin.

273 citations


Journal ArticleDOI
TL;DR: Endocrine system is involved in the control of ejaculatory function and that PRL, TSH and testosterone play an independent role, according to a large series of subjects consulting for sexual dysfunction.
Abstract: Although it is well established that all the aspects of male reproduction are hormonally regulated, the endocrine control of the ejaculatory reflex is still not completely clarified. Sex steroids, thyroid and pituitary hormones (oxytocin and prolactin) have been proposed to control the ejaculatory process at various levels; however, only a few reports are currently available. The aim of this study was to evaluate the contribution of testosterone, thyrotropin (TSH) and prolactin (PRL) in the pathogenesis of ejaculatory dysfunction in a large series of subjects consulting for sexual dysfunction. Among the 2652 patients studied, 674 (25.2%) and 194 (7.3%) reported premature and delayed ejaculation (PE and DE), respectively. Categorizing ejaculatory difficulties on an eight-point scale starting from severe PE and ending with anejaculation (0 = severe PE, 1 = moderate PE, 2 = mild PE, 3 = no difficulties, 4 = mild DE, 5 = moderate DE, 6 = severe DE and 7 = anejaculation), PRL as well as TSH levels progressively increased from patients with severe PE towards those with anejaculation. Conversely, the opposite was observed for testosterone levels. All of these associations were confirmed after adjustment for age (adjusted r = 0.050, 0.053 and -0.038 for PRL, TSH and testosterone, respectively; all p < 0.05). When all hormonal parameters were introduced in the same regression model, adjusting for age, ΣMHQ (an index of general psychopathology) and use of selective serotonin reuptake inhibitor antidepressants, they were independently associated with ejaculatory problems (adjusted r = 0.056, 0.047 and -0.059 for PRL, TSH and testosterone, respectively; all p < 0.05). This study indicates endocrine system is involved in the control of ejaculatory function and that PRL, TSH and testosterone play an independent role.

133 citations


Journal ArticleDOI
TL;DR: Prolactin does increase in response to psychosocial stress, however, with large individual variation in magnitude of response, indicating that individual differences in prolactin response in healthy men and women are dependent on the general physiological stress activation.

121 citations


Journal ArticleDOI
TL;DR: Data from dual-label in situ hybridization investigated whether prolactin-receptor (PRLR) mRNA is expressed in GnRH neurons, finding that it was highly expressed throughout the rostral preoptic area and there was a high degree of colocalization of PRLR mRNA in both Gad1/Gad2 and Kiss1 mRNA-containing cells.
Abstract: Prolactin receptor mRNA is expressed in GABA and kisspeptin neurons in the rostral hypothalamus but in relatively few GnRH neurons, suggesting that hyperprolactinemia might cause infertility through actions on afferent inputs to GnRH neurons.

103 citations


Journal ArticleDOI
TL;DR: There is a need for a randomised controlled trial of low dose dopamine agonist therapy versus sex steroid replacement to establish the relative safety and efficacy of each approach.
Abstract: Background and objectives: Antipsychotic medications are a potential cause of hyperprolactinaemia and may be implicated in the development of pituitary adenomas. This review examines the effect of different antipsychotic medications on prolactin and sexual function, and provides practical guidelines for investigation and management of antipsychotic-induced hyperprolactinaemia.Method: Literature review.Results and conclusions: Antipsychotic-induced hyperprolactinaemia occurs overall in up to 70% of patients with schizophrenia, depending on the medications used. It is associated with significant levels of hypogonadism and sexual dysfunction, which in general relates to the degree of prolactin elevation. A consequence of the hypogonadism is clinically significant bone loss which may lead to osteoporosis and increased risk of minimal trauma fracture. Where the potentially offending drug cannot be safely withdrawn to document a normal prolactin, imaging with MRI should be undertaken to exclude a structural pit...

102 citations


Journal ArticleDOI
TL;DR: The remarkable variability in the clinical presentation of affected patients indicates the influence of the genetic background, environmental factors and possibly stochastic events of this heterogeneous disease remains unexplained, which further suggests the involvement of additional genes.

97 citations


Journal ArticleDOI
TL;DR: Although plasma OT did not increase reliably from pre-stress levels during stressors, greater overall OT level was related to greater vasodilation and cardiac stroke volume responses to both tasks, and higher NE was linked to increases in heart rate and decreases in stroke volume.

92 citations


Journal ArticleDOI
TL;DR: Few controlled studies of dopamine agonist treatment in humans with autoimmune disease have been conducted only in systemic lupus erythematosus patients, which support the potential efficacy of such agents even during pregnancy and postpartum.
Abstract: The relationship between prolactin and the immune system has been demonstrated in the last two decades, opening new windows in the field of the immunoendocrinology. Prolactin has an important role in the innate and adaptive immune response. Increased prolactin levels have been described in autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, Sjogren syndrome, and systemic sclerosis among others. Hyperprolactinemia is associated with active disease and organ involvement in systemic lupus erythematosus. Therefore, prolactin is an integral member of the immunoneuroendocrinology network and seems to have a role in pathogenesis of autoimmune diseases. Few controlled studies of dopamine agonist treatment in humans with autoimmune disease have been conducted only in systemic lupus erythematosus patients, which support the potential efficacy of such agents even during pregnancy and postpartum. Further studies are necessary to elucidate the mechanisms by which prolactin affects autoimmune disease activity, increase the inflammatory mechanism, and determine the role of anti-prolactinemic drugs to regulate the immune/inflammatory process.

88 citations


Journal ArticleDOI
01 Jul 2011-Steroids
TL;DR: Further data are required, with better assays and repeat measures, to provide more accurate estimates of risk and to clarify the role of oestrogens in premenopausal women and the roles of other endogenous hormones.

87 citations


Journal ArticleDOI
TL;DR: The data are the first to provide evidence that, in a primate model, there is a functional Kp-signaling system within the pituitary, which is dynamically regulated and may contribute to the direct control of gonadotropic and somatotropic axes.
Abstract: Kisspeptins (Kps) have emerged as key players in the control of reproductive-axis function, in which they operate as primary regulators of hypothalamic GnRH release. In addition, recent data indicate that Kps can also directly act on the pituitary to stimulate LH and GH release in primary pituitary cell culture prepared from rats, cows, and sheep. We present herein evidence that Kps (specifically Kp-10) can also stimulate LH and GH release in primary pituitary cell cultures prepared from female baboons (Papio anubis), a species that more closely models human physiology. The stimulatory effect of Kp-10 on LH and GH release was dose and time dependent and enhanced the hormonal responses to their major regulators (GnRH for LH; GHRH/ghrelin for GH) without affecting the release of other pituitary hormones (TSH, FSH, ACTH, prolactin). Use of pharmacological intracellular signaling blockers indicated Kp-10 signals through phospholipase C, protein kinase C, MAPK, and intracellular Ca2+ mobilization, but not adenylyl cyclase, protein kinase A, extracellular Ca2+ influx (through L-type channels), or nitric oxide synthase, to stimulate both LH and GH release. Interestingly, blockade of mammalian target of rapamycin or phosphoinositol 3-kinase activity fully abolished the stimulatory effect of Kp-10 on LH but not GH release. Of note, estradiol enhanced the relative LH response to Kp-10, alone or in combination with GnRH. In sum, our data are the first to provide evidence that, in a primate model, there is a functional Kp-signaling system within the pituitary, which is dynamically regulated and may contribute to the direct control of gonadotropic and somatotropic axes.

Journal ArticleDOI
TL;DR: Investigation of 3 cases of adult-onset combined GH, PRL, and TSH deficiencies and found that the endocrinological phenotype in each was linked to autoimmunity directed against the PIT-1 protein, suggesting a unique "anti-PIT-1 antibody syndrome," related to APS.
Abstract: The pituitary-specific transcriptional factor-1 (PIT-1, also known as POU1F1), is an essential factor for multiple hormone-secreting cell types. A genetic defect in the PIT-1 gene results in congenital growth hormone (GH), prolactin (PRL), and thyroid-stimulating hormone (TSH) deficiency. Here, we investigated 3 cases of adult-onset combined GH, PRL, and TSH deficiencies and found that the endocrinological phenotype in each was linked to autoimmunity directed against the PIT-1 protein. We detected anti-PIT-1 antibody along with various autoantibodies in the patients' sera. An ELISA-based screening revealed that this antibody was highly specific to the disease and absent in control subjects. Immunohistochemical analysis revealed that PIT-1-, GH-, PRL-, and TSH-positive cells were absent in the pituitary of patient 2, who also had a range of autoimmune endocrinopathies. These clinical manifestations were compatible with the definition of autoimmune polyendocrine syndrome (APS). However, the main manifestations of APS-I--hypoparathyroidism and Candida infection--were not observed and the pituitary abnormalities were obviously different from the hypophysitis associated with APS. These data suggest that these patients define a unique "anti-PIT-1 antibody syndrome," related to APS.

Journal ArticleDOI
TL;DR: New insight is provided into the molecular and cellular targets by which locally produced prolactin could contribute to prostate cancer initiation and progression and a number of pharmacological inhibitors targeting various levels of the PRLR–Jak2–Stat5a/b pathway have been developed and are entering clinical trials for advanced prostate cancer.
Abstract: Prolactin is best known for its actions on the mammary gland. However, circulating prolactin is also detected in males and its receptor (PRLR) is expressed in the prostate, suggesting that the prostate is a target of prolactin. Germline knockout of prolactin or its receptor has failed to reveal a key role for prolactin signaling in mouse prostate physiology. However, several studies involving rodent models and human prostate cell lines and specimens have supported the contribution of the canonical PRLR-Jak2-Stat5a/b pathway to prostate cancer tumorigenesis and progression. Increased expression of prolactin in the prostate itself (rather than changes in circulating prolactin levels) and crosstalk with androgen receptor (AR) signaling are potential mechanisms for increased Stat5a/b signaling in prostate cancer. In the mouse prostate, prolactin overexpression results in disorganized expansion of the basal/stem cell compartment, which has been proposed to house putative prostate tumor-initiating cells. These findings provide new insight into the molecular and cellular targets by which locally produced prolactin could contribute to prostate cancer initiation and progression. A number of pharmacological inhibitors targeting various levels of the PRLR-Jak2-Stat5a/b pathway have been developed and are entering clinical trials for advanced prostate cancer.

Journal ArticleDOI
19 Jan 2011-PLOS ONE
TL;DR: The results suggest the age-related decline in GH helps to preserve systemic insulin sensitivity, and in the context of moderate caloric intake, prevents the deterioration in metabolic function.
Abstract: Growth hormone (GH) inhibits fat accumulation and promotes protein accretion, therefore the fall in GH observed with weight gain and normal aging may contribute to metabolic dysfunction. To directly test this hypothesis a novel mouse model of adult onset-isolated GH deficiency (AOiGHD) was generated by cross breeding rat GH promoter-driven Cre recombinase mice (Cre) with inducible diphtheria toxin receptor mice (iDTR) and treating adult Cre+/−,iDTR+/− offspring with DT to selectively destroy the somatotrope population of the anterior pituitary gland, leading to a reduction in circulating GH and IGF-I levels. DT-treated Cre−/−,iDTR+/− mice were used as GH-intact controls. AOiGHD improved whole body insulin sensitivity in both low-fat and high-fat fed mice. Consistent with improved insulin sensitivity, indirect calorimetry revealed AOiGHD mice preferentially utilized carbohydrates for energy metabolism, as compared to GH-intact controls. In high-fat, but not low-fat fed AOiGHD mice, fat mass increased, hepatic lipids decreased and glucose clearance and insulin output were impaired. These results suggest the age-related decline in GH helps to preserve systemic insulin sensitivity, and in the context of moderate caloric intake, prevents the deterioration in metabolic function. However, in the context of excess caloric intake, low GH leads to impaired insulin output, and thereby could contribute to the development of diabetes.

Journal ArticleDOI
TL;DR: Targeting metabolic pathways that are governed by PRL, which has already been implicated in the progression of breast cancer, may be of therapeutic benefit, and stimulating CPT1 expression and/or activity in MCF-7 and MDA-MB-231 breast cancer cells is investigated.
Abstract: Background Mammary carcinomas have been associated with a high-fat diet, and the rate of breast cancer in overweight post-menopausal women is up to 50% higher than in their normal-weight counterparts. Epidemiological studies suggest that prolactin (PRL) plays a role in the progression of breast cancer. The current study examined breast cancer as a metabolic disease in the context of altered fatty acid catabolism by examining the effect of PRL on carnitine palmitoyl transferase 1 (CPT1), an enzyme that shuttles long-chain fatty acids into the mitochondrial matrix for β-oxidation. The effect of PRL on the adenosine 5'-monophosphate-activated protein kinase (AMPK) energy sensing pathway was also investigated.

Journal ArticleDOI
01 Sep 2011-Peptides
TL;DR: This minireview summarizes the recent data about the action of tachykinins on the hypothalamo-pituitary-gonadal axis and indicates that it may play an important role in the ovarian function.

Journal ArticleDOI
TL;DR: It is demonstrated that prolactin can promote diverse carcinomas in mice, many of which resemble luminal breast cancers, providing a novel experimental model to examine the pathogenesis, progression and treatment responsiveness of this tumor subtype.
Abstract: Tumors that express estrogen receptor alpha (ERα+) comprise 75% of breast cancers in women. While treatments directed against this receptor have successfully lowered mortality rates, many primary tumors initially or later exhibit resistance. The paucity of murine models of this "luminal" tumor subtype has hindered studies of factors that promote their pathogenesis and modulate responsiveness to estrogen-directed therapeutics. Since epidemiologic studies closely link prolactin and the development of ERα+ tumors in women, we examined characteristics of the aggressive ERα+ and ERα- carcinomas which develop in response to mammary prolactin in a murine transgenic model (neu-related lipocalin- prolactin (NRL-PRL)). To evaluate their relationship to clinical tumors, we determined phenotypic relationships among these carcinomas, other murine models of breast cancer, and features of luminal tumors in women. We examined a panel of prolactin-induced tumors for characteristics relevant to clinical tumors: histotype, ERα/progesterone receptor (PR) expression and estrogen responsiveness, Activating Protein 1 (AP-1) components, and phosphorylation of signal transducer and activator of transcription 5 (Stat5), extracellular signal regulated kinase (ERK) 1/2 and AKT. We compared levels of transcripts in the ERα-associated "luminal" signature that defines this subtype of tumors in women and transcripts enriched in various mammary epithelial lineages to other well-studied genetically modified murine models of breast cancer. Finally, we used microarray analyses to compare prolactin-induced ERα+ and ERα- tumors, and examined responsiveness to estrogen and the anti-estrogen, Faslodex, in vivo. Prolactin-induced carcinomas were markedly diverse with respect to histotype, ERα/PR expression, and activated signaling cascades. They constituted a heterogeneous, but distinct group of murine mammary tumors, with molecular features of the luminal subtype of human breast cancer. In contrast to morphologically normal and hyperplastic structures in NRL-PRL females, carcinomas were insensitive to ERα-mediated signals. These tumors were distinct from mouse mammary tumor virus (MMTV)-neu tumors, and contained elevated transcripts for factors associated with luminal/alveolar expansion and differentiation, suggesting that they arose from physiologic targets of prolactin. These features were shared by ERα+ and ERα- tumors, suggesting a common origin, although the former exhibited transcript profiles reflecting greater differentiation. Our studies demonstrate that prolactin can promote diverse carcinomas in mice, many of which resemble luminal breast cancers, providing a novel experimental model to examine the pathogenesis, progression and treatment responsiveness of this tumor subtype.

Journal ArticleDOI
TL;DR: It is determined that chronic intraperitoneal injections of different dosages of prolactin have opposite effects on insulin resistance and β‐cell function and mass in 90% pancreatectomized diabetic male rats, and the mechanisms were explored.
Abstract: Background Prolactin improves glucose homeostasis by increasing β-cell mass under certain conditions such as pregnancy, whereas hyperprolactinaemia due to a pituitary gland adenoma tumour exacerbates insulin resistance. However, previous studies have not evaluated how prolactin modulates β-cell function and insulin sensitivity at different dosages. Here, we determined that chronic intraperitoneal injections of different dosages of prolactin have opposite effects on insulin resistance and β-cell function and mass in 90% pancreatectomized diabetic male rats, and the mechanisms were explored. Methods Diabetic rats were divided into three groups according to the dose of intraperitoneally injected prolactin for 4 weeks: (1) low dose of prolactin (25 µg/kg bw/12 h), (2) high dose of prolactin (250 µg/kg bw/12 h), and (3) vehicle. As a non-diabetic control group, sham-operated rats were injected with vehicle. Results Chronic high- and low-dose prolactin injections elevated serum prolactin levels by 2.5- and 11.8-fold, respectively. Both dosages promoted β-cell mass by increasing β-cell proliferation and neogenesis through the potentiation of phosphorylation of signal transducer and activator of transcription 5 and decreased menin expression in diabetic rats. However, only the low-dose prolactin injection potentiated glucose-stimulated insulin secretion though glucokinase and glucose transporter 2 induction in the diabetic rats. In addition, low-dose prolactin decreased hepatic glucose output in hyperinsulinaemic states, indicating an improvement in hepatic insulin resistance. However, the high-dose prolactin injection exacerbated whole-body and hepatic insulin resistance in diabetic rats. Conclusions In contrast to the normal adaptive increases in glucose-stimulated insulin secretion through expanded β-cell mass and insulin sensitivity realized with moderately increased prolactin levels, high levels of prolactin exacerbate insulin resistance and impair the insulin-secretory capacity in diabetic mice. Copyright © 2011 John Wiley & Sons, Ltd.

Journal ArticleDOI
TL;DR: Cortistatin deficit uncovered a gender-dependent role of this peptide in the regulation of glucose-insulin homeostasis, because male, but not female, cort-/- mice developed insulin resistance.
Abstract: Cortistatin (CST) and somatostatin (SST) evolve from a common ancestral gene and share remarkable structural, pharmacological, and functional homologies. Although CST has been considered as a natural SST-analogue acting through their shared receptors (SST receptors 1–5), emerging evidence indicates that these peptides might in fact exert unique roles via selective receptors [e.g. CST, not SST, binds ghrelin receptor growth hormone secretagogue receptor type 1a (GHS-R1a)]. To determine whether the role of endogenous CST is different from SST, we characterized the endocrine-metabolic phenotype of male/female CST null mice (cort−/−) at hypothalamic-pituitary-systemic (pancreas-stomach-adrenal-liver) levels. Also, CST effects on hormone expression/secretion were evaluated in primary pituitary cell cultures from male/female mice and female primates (baboons). Specifically, CST exerted an unexpected stimulatory role on prolactin (PRL) secretion, because both male/female cort−/− mice had reduced PRL levels, and CST treatment (in vivo and in vitro) increased PRL secretion, which could be blocked by a GHS-R1a antagonist in vitro and likely relates to the decreased success of female cort−/− in first-litter pup care at weaning. In contrast, CST inhibited GH and adrenocorticotropin-hormone axes in a gender-dependent fashion. In addition, a rise in acylated ghrelin levels was observed in female cort−/− mice, which were associated with an increase in stomach ghrelin/ghrelin O-acyl transferase expression. Finally, CST deficit uncovered a gender-dependent role of this peptide in the regulation of glucose-insulin homeostasis, because male, but not female, cort−/− mice developed insulin resistance. The fact that these actions are not mimicked by SST and are strongly gender dependent offers new grounds to investigate the hitherto underestimated physiological relevance of CST in the regulation of physiological/metabolic processes.

Journal ArticleDOI
TL;DR: This review focuses on treatment options for aggressive pituitary tumors, including surgery, radiotherapy and medical treatment, as well as focusing on the promising therapeutic options, evaluating the literature of the last 15 years.
Abstract: Introduction: Aggressive tumors of the pituitary gland are classically defined as pituitary tumors with a massive invasion of the surrounding anatomical structures and rapid growth. They are notoriously difficult to manage and are associated with poor prognosis because the therapeutic options are limited and the tumors are generally unresponsive to therapy. Areas covered: This review focuses on treatment options for aggressive pituitary tumors, including surgery, radiotherapy and medical treatment, as well as focusing on the promising therapeutic options for aggressive pituitary tumors, evaluating the literature of the last 15 years. With the exception of prolactinomas, surgery is the first-line option, but most aggressive pituitary tumors often require repeated surgery. Pharmacotherapies are useful when surgery is unlikely to improve symptoms, or as an adjunct therapy to surgery. In prolactinomas, dopamine agonists are the first-line treatment and normalize prolactin levels in most patients, even those w...

Journal ArticleDOI
TL;DR: Investigation of potential signaling effects of both hormones in the estrogen receptor- and progesterone receptor-positive human T47D breast cancer cell line found that this cell type expresses ample GHR and PRLR and responds well to both hGH and hPRL, broadening the understanding of how these related hormones and their related receptors may function in physiology and pathophysiology.
Abstract: GH receptor (GHR) and prolactin (PRL) receptor (PRLR) are structurally similar cytokine receptor superfamily members that are highly conserved among species. GH has growth-promoting and metabolic effects in various tissues in vertebrates, including humans. PRL is essential for regulation of lactation in mammals. Recent studies indicate that breast tissue bears GHR and PRLR and that both GH and PRL may impact development or behavior of breast cancer cells. An important facet of human GH (hGH) and human PRL (hPRL) biology is that although hPRL interacts only with hPRLR, hGH binds well to both hGHR and hPRLR. Presently, we investigated potential signaling effects of both hormones in the estrogen receptor- and progesterone receptor-positive human T47D breast cancer cell line. We found that this cell type expresses ample GHR and PRLR and responds well to both hGH and hPRL, as evidenced by activation of the Janus kinase 2/signal transducer and activator of transcription 5 pathway. Immunoprecipitation studies revealed specific GHR-PRLR association in these cells that was acutely enhanced by GH treatment. Although GH caused formation of disulfide-linked and chemically cross-linked GHR dimers in T47D cells, GH preferentially induced tyrosine phosphorylation of PRLR rather than GHR. Notably, both a GHR-specific ligand antagonist (B2036) and a GHR-specific antagonist monoclonal antibody (anti-GHR(ext-mAb)) failed to inhibit GH-induced signal transducer and activator of transcription 5 activation. In contrast, although the non-GHR-specific GH antagonist (G120R) and the PRL antagonist (G129R) individually only partially inhibited GH-induced activation, combined treatment with these two antagonists conferred greater inhibition than either alone. These data indicate that endogenous GHR and PRLR associate (possibly as a GHR-PRLR heterodimer) in human breast cancer cells and that GH signaling in these cells is largely mediated by the PRLR in the context of both PRLR-PRLR homodimers and GHR-PRLR heterodimers, broadening our understanding of how these related hormones and their related receptors may function in physiology and pathophysiology.

Journal ArticleDOI
TL;DR: A new role is established for the FGFR4-G388R polymorphism in pituitary oncogenesis, providing a rationale for targeting Src and STAT3 in the personalized treatment of associated disorders.
Abstract: Pituitary tumors are common intracranial neoplasms, yet few germline abnormalities have been implicated in their pathogenesis. Here we show that a single nucleotide germline polymorphism (SNP) substituting an arginine (R) for glycine (G) in the FGFR4 transmembrane domain can alter pituitary cell growth and hormone production. Compared with FGFR4-G388 mammosomatotroph cells that support prolactin (PRL) production, FGFR4-R388 cells express predominantly growth hormone (GH). Growth promoting effects of FGFR4-R388 as evidenced by enhanced colony formation was ascribed to Src activation and mitochondrial serine phosphorylation of STAT3 (pS-STAT3). In contrast, diminished pY-STAT3 mediated by FGFR4-R388 relieved GH inhibition leading to hormone excess. Using a knock-in mouse model, we demonstrate the ability of FGFR4-R385 to promote GH pituitary tumorigenesis. In patients with acromegaly, pituitary tumor size correlated with hormone excess in the presence of the FGFR4-R388 but not the FGFR4-G388 allele. Our findings establish a new role for the FGFR4-G388R polymorphism in pituitary oncogenesis, providing a rationale for targeting Src and STAT3 in the personalized treatment of associated disorders.

Journal ArticleDOI
TL;DR: It is concluded that the seasonal change in kisspeptin neurones cannot be regulated directly by melatonin, although it may be a result of changes in prolactin levels.
Abstract: Melatonin is secreted at night by the pineal gland and governs the reproductive system in seasonal breeders, such as sheep. The mechanism by which melatonin regulates reproduction is not known. The circannual rhythmicity of other factors, including prolactin, is also regulated by photoperiod via changes in melatonin secretion. In sheep, plasma prolactin levels are higher in the nonbreeding season than the breeding season. Kisspeptin, synthesised by neurones in the ovine arcuate nucleus (ARC) and preoptic area, is a key regulator of reproduction through stimulation of gonadotrophin-releasing hormone secretion and its expression in the ARC is reduced during the nonbreeding season. We hypothesised that kisspeptin expression is directly, or indirectly, regulated by melatonin and/or prolactin. We first examined the expression of melatonin receptor (MTNR1A) in kisspeptin (Kiss1 mRNA) neurones in the ARC of ovariectomised (OVX) sheep using double-label in situ hybridisation. MTNR1A mRNA was not expressed by kisspeptin neurones, whereas strong expression was detected in the pars tuberalis. We then examined the expression of the long-form prolactin receptor (PRLR-L) in ARC kisspeptin neurones. In OVX ewes, approximately 60% of kisspeptin neurones expressed PRLR-L mRNA at similar levels in the breeding and nonbreeding seasons. We then aimed to determine whether prolactin treatment during the breeding season regulates kisspeptin expression in the ARC. Continuous central infusion of prolactin (20 μg/h for 7 days) in oestradiol-treated OVX sheep did not alter Kiss1 mRNA expression or luteinising hormone secretion, although it induced substantial phosphorylated signal transducer and activator of transcription 5-immunoreactive nuclei staining in the mediobasal hypothalamus. We conclude that the seasonal change in kisspeptin neurones cannot be regulated directly by melatonin, although it may be a result of changes in prolactin levels. Despite this, kisspeptin expression was unchanged after exogenous prolactin treatment in breeding season ewes.

Journal ArticleDOI
TL;DR: During pregnancy, placental hormones act through the prolactin receptor to increase β-cell mass by up regulatingβ-cell proliferation by engaging Jak2, Akt, menin/p18, and p21.
Abstract: Pregnancy-induced increase in β-cell proliferation requires intact prolactin receptors, an increase involving up-regulation of Jak2, IRS-2, Akt, and p21 expression and suppression of menin/p18 pathways.

Journal ArticleDOI
TL;DR: In this article, the authors investigated levels of functional prolactin-mediated signal transduction in the brain of lactating mice, using Prolactininduced phosphorylation of signal transducer and activator of transcription 5 (pSTAT5) as a marker.
Abstract: During lactation, there are numerous functional adaptations in the maternal brain. There is evidence that the high levels of circulating prolactin present during lactation might contribute to these adaptive changes. The present study aimed to investigate levels of functional prolactin-mediated signal transduction in the brain of lactating mice, using prolactin-induced phosphorylation of signal transducer and activator of transcription 5 (pSTAT5) as a marker, and compare these to the effect of exogenous prolactin during diestrus. On Day 7 of lactation, widespread induction of pSTAT5 was observed in numerous regions of the mouse forebrain and brainstem. In the medial preoptic nucleus, bed nuclei stria terminalis, paraventricular nucleus, and medial amygdala of the forebrain, and in the rostral periaqueductal gray, parabrachial nucleus, dorsal raphe, and the raphe obscurus nucleus of the brainstem, pSTAT5 expression was markedly increased during lactation compared with the response to exogenous prolactin during diestrus. In the anteroventral periventricular nucleus, arcuate nucleus, ventromedial nucleus, and dorsomedial nucleus, responses in lactation were comparable to diestrus. Conversely, in the area postrema of the brainstem, there was a reduction in response to prolactin, with a loss of pSTAT5 expression, during lactation. These differential responses following either acute or chronic elevations in prolactin were not accompanied by any changes in levels of prolactin receptor mRNA, when measured by in situ hybridization. These data are consistent with the hypothesis that prolactin might mediate widespread adaptive responses in the maternal brain.

Journal ArticleDOI
TL;DR: This is the first evidence indicating that antipsychotic drug treatment in men is associated with a variant in the DRD2 gene in which the -141C Del allele might be a protective factor.
Abstract: Aim: Sexual dysfunction induced by antipsychotic drug treatment is under investigated and under reported. This study aimed to determine the influence of genetic polymorphisms in the D2 dopamine receptor ( DRD2) and endothelial nitric oxide synthase (eNOS) genes, and the possible role of blood prolactin concentrations on sexual function in schizophrenic patients. Materials & methods: Male remitted schizophrenic patients (n = 100), who were living with a sexual partner and receiving antipsychotic drug monotherapy for at least 6 months, were assessed for sexual and erectile dysfunction using the Arizona Sexual Experience Scale and the five-item version of the International Index of Erectile Function. Blood samples were taken for plasma prolactin determination and genotyped for four polymorphisms: DRD2 (-141C Ins/Del and Taq1A) and eNOS gene (G894T and T-786C). Results: The -141C Ins/Del, but not Taq1A, polymorphism of the DRD2 gene was significantly associated with sexual dysfunction with the del allele being less frequent in sexual dysfunction subjects. Neither of the eNOS polymorphisms, G894T or T-786C, was significantly associated with sexual or erectile dysfunction. Prolactin concentrations were significantly higher in patients with erectile dysfunction but did not reach significance in those with sexual dysfunction. Prolactin was also reduced in -141C Del allele carriers. The frequency and severity of sexual dysfunction in the patients receiving typical antipsychotics was significantly greater than those receiving risperidone or clozapine, while prolactin concentrations were significantly higher in subjects receiving risperidone compared with those receiving clozapine or typical antipsychotics. Conclusion: This is the first evidence indicating that antipsychotic drug treatment in men is associated with a variant in the DRD2 gene in which the -141C Del allele might be a protective factor. While this may, in part, be mediated by effects on prolactin, other factors are likely to contribute to the greater sexual dysfunction in patients receiving typical antipsychotics. Original submitted 25 January 2011; Revision submitted 21 March 2011


Journal ArticleDOI
TL;DR: The remission rates of male patients with microadenomas and/or intrasellar macroprolactinomas by surgery alone remain excellent, and surgery followed by adjuvant therapy as necessary is required for optimizing management of male prolactinoma, especially for extrasellar macroadenomas.
Abstract: Context: Little systematic data on male prolactinomas treated with surgery are available. Objective: To clarify the clinical features and confirm the efficacy of transsphenoidal surgery for male prolactinomas and predictive factors after initial surgery. Patients and methods: This retrospective study included 87 male patients with prolactinoma treated by transsphenoidal surgery at an academic medical center. Hormonal and visual status, remission rates, and the rate of tumor relapse, as well as predictive factors, were evaluated. Results: Postoperative initial remission was achieved in 52.9% of patients. The remission rate was markedly higher in microadenomas (83.3%) than in macroadenomas (44.9%). Logistic regression analysis showed that the predictive factors of the early negative outcomes were high preoperative prolactin (PRL) levels and tumor invasion. After a median follow-up of 45 months, the long-term remission rate was 42.5%, and relapse of hyperprolactinemia occurred in 19.6% of the cured patients. The 5-year recurrence-free survival was 78.2% (95% confidence interval, 62.3‐88.1%). When surgery was followed by adjuvant treatment in uncured and recurrent patients, 78.8% of patients in the entire group in the absence of dopamine agonists obtained biochemical remission at the end of follow-up. Conclusion: Transsphenoidal surgery is a viable treatment alternative for male prolactinomas. The remission rates of male patients with microadenomas and/or intrasellar macroprolactinomas by surgery alone remain excellent, and surgery followed by adjuvant therapy as necessary is required for optimizing management of male prolactinomas, especially for extrasellar macroprolactinomas. The early negative results are associated with preoperative PRL levels and tumor invasion.

Journal ArticleDOI
01 Jun 2011-Peptides
TL;DR: It is suggested that increased serum prolactin and nesfatin-1 concentrations, decreased ghrelin concentrations could be used as markers to identify patients that have suffered a recent epileptic seizure or other paroxysmal event (psychogenic).

Journal ArticleDOI
TL;DR: The findings demonstrate that the prolactin receptor is present in macrophages of the atherosclerotic plaque at sites of most prominent inflammation, and proposes that prolACTin receptor signaling contributes to the local inflammatory response within the Atherosclerosis plaque and thus to atherogenesis.
Abstract: Atherosclerotic vascular disease is the consequence of a chronic inflammatory process, and prolactin has been shown to be a component of the inflammatory response. Additionally, recent studies indicate that prolactin contributes to an atherogenic phenotype. We hypothesized that this may be the result of a direct effect of prolactin on atherogenesis through activation of the prolactin receptor. Human carotid atherosclerotic plaques were obtained from patients by endarteriectomies. The mRNA of prolactin receptor, but not of prolactin, was detected in these atherosclerotic plaques by quantitative realtime PCR. In situ hybridization confirmed the expression of the prolactin receptor in mononuclear cells. Analysis at the protein level using immunohistochemistry and immunoelectron microscopy revealed that the prolactin receptor was abundantly present in macrophages near the lipid core and shoulder regions of the plaques. Our findings demonstrate that the prolactin receptor is present in macrophages of the atherosclerotic plaque at sites of most prominent inflammation. We therefore propose that prolactin receptor signaling contributes to the local inflammatory response within the atherosclerotic plaque and thus to atherogenesis.