Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

No effect of unacylated ghrelin administration on subcutaneous PC3 xenograft growth or metabolic parameters in a Rag1-/- mouse model of metabolic dysfunction

  • Michelle L. Maugham,

    Roles Conceptualization, Data curation, Formal analysis, Investigation, Methodology, Validation, Writing – original draft, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Skeletal Biology and Forensic Anthropology Research Laboratory, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia

  • Inge Seim,

    Roles Conceptualization, Funding acquisition, Methodology, Supervision, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing, China

  • Patrick B. Thomas,

    Roles Investigation, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Gabrielle J. Crisp,

    Roles Investigation, Methodology, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Esha T. Shah,

    Roles Investigation, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Adrian C. Herington,

    Roles Funding acquisition, Methodology, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Kristy A. Brown,

    Roles Conceptualization, Methodology, Writing – review & editing

    Affiliation Department of Medicine, Weill Cornell Medicine, New York City, New York, United States of America

  • Laura S. Gregory,

    Roles Supervision, Writing – review & editing

    Affiliation Skeletal Biology and Forensic Anthropology Research Laboratory, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia

  • Colleen C. Nelson,

    Roles Funding acquisition, Project administration, Resources, Writing – review & editing

    Affiliation Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Penny L. Jeffery,

    Roles Conceptualization, Investigation, Methodology, Project administration, Supervision, Visualization, Writing – review & editing

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

  • Lisa K. Chopin

    Roles Conceptualization, Funding acquisition, Methodology, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    l.chopin@qut.edu.au

    Affiliations Ghrelin Research Group, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Australian Prostate Cancer Research Centre - Queensland, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia, Comparative and Endocrine Biology Laboratory, Translational Research Institute – Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia

Abstract

Ghrelin is a peptide hormone which, when acylated, regulates appetite, energy balance and a range of other biological processes. Ghrelin predominately circulates in its unacylated form (unacylated ghrelin; UAG). UAG has a number of functions independent of acylated ghrelin, including modulation of metabolic parameters and cancer progression. UAG has also been postulated to antagonise some of the metabolic effects of acyl-ghrelin, including its effects on glucose and insulin regulation. In this study, Rag1-/- mice with high-fat diet-induced obesity and hyperinsulinaemia were subcutaneously implanted with PC3 prostate cancer xenografts to investigate the effect of UAG treatment on metabolic parameters and xenograft growth. Daily intraperitoneal injection of 100 μg/kg UAG had no effect on xenograft tumour growth in mice fed normal rodent chow or 23% high-fat diet. UAG significantly improved glucose tolerance in host Rag1-/- mice on a high-fat diet, but did not significantly improve other metabolic parameters. We propose that UAG is not likely to be an effective treatment for prostate cancer, with or without associated metabolic syndrome.

Introduction

The peptide hormone ghrelin is a circulating appetite-stimulating hormone which regulates a number of other biological processes [13]. These include metabolism and energy balance [14], and diseases such as cancer [5]. Ghrelin acts via its cognate receptor, the growth hormone secretagogue receptor 1a (GHSR1a), a G protein-coupled receptor [6], and one or more unknown alternative receptors [710]. In order to activate GHSR1a at physiological concentrations, ghrelin must be acylated at its third residue, a serine [11, 12], by the enzyme ghrelin O-acyl transferase (GOAT) [11, 12].

The major circulating form of ghrelin is its unmodified form, unacylated ghrelin (UAG; also known as DAG). UAG, which does not directly stimulate feeding [5], was initially considered to be functionally inactive, but is now appreciated to bind to and activate a distinct, unknown receptor [4, 1318] and have a number of functions [1922]. UAG plays roles in the regulation of glucose and energy balance and has effects on cell proliferation [1923]. Importantly, it may oppose some of the effects of acyl-ghrelin [16, 2426] by preventing the rise in circulating glucose and insulin associated with acyl-ghrelin administration in rodents [22, 26, 27]. From these studies, it is apparent that UAG is an endocrine hormone in its own right [20]. UAG and the truncated, cyclised UAG analogue AZP-531 prevented the development of pre-diabetes in C57BL/6 mice fed a high-fat diet for two weeks, highlighting a potential of unacylated forms of ghrelin as treatments for metabolic syndrome [27]. In human trials, UAG had similar effects, improving glycaemic control and insulin sensitivity in patients with type 2 diabetes mellitus [28] and improving glucose handling and reducing free fatty acids in healthy subjects when administered overnight as a continuous infusion [29]. AZP-531 also had beneficial effects on glucose balance and led to weight loss in patients with type 2 diabetes mellitus in a phase I clinical trial [30]. Similar benefits have been observed in patients with Prader-Willi syndrome, a genetic disorder associated with hyperghrelinaemia and obesity [31].

Close to two decades of work has firmly established a role for the ghrelin axis in cancer [5, 3235]. This includes prostate cancer, a classical endocrine-related cancer and the most commonly diagnosed cancer in American men after skin cancer [36], where acyl-ghrelin increases cell proliferation and migration [5, 14, 3746]. UAG also has functional effects in several cancers, including prostate cancer [5, 3234, 43]. In the PC3 prostate cancer cell line UAG has a biphasic effect, reducing cell proliferation at supraphysiological levels (10nM-1μM) [14].

Studies investigating the role of UAG in prostate cancer have been limited to in vitro experiments. In vivo studies are required, however. Obesity, overweight, and co-morbidities (including hyperinsulinaemia) are now recognised as critical risk factors for numerous cancers [4749]. These include cancer types with high-prevalence and mortality, such as tumours of the prostate, endometrium, breast, and gastrointestinal system [4755]. Obesity and increased body mass have been associated with increased risk of advanced prostate cancer, more aggressive and high-grade disease, and increased risk of death from prostate cancer [5659]. Castration-resistant prostate cancer (CRPC) occurs when prostate cancer recurs after remission from androgen-targeted therapies (ATT) [60]. Treatments for CRPC are limited and this stage of the disease often results in the formation of painful, metastatic bone lesions and associated morbidity and mortality [6163]. Metabolic syndrome and hyperinsulinaemia are common side effects of ATT [64, 65] and may also further accelerate the progression to CRPC [48, 58, 6668]. As UAG reduces prostate cancer proliferation in vitro [14] and has potential beneficial metabolic effects in vivo, we examined the effect of UAG in our model of metabolic dysfunction: Rag1-/- mice fed a high-fat diet with subcutaneous prostate cancer cell line xenografts [69].

Materials and methods

Cell culture

Human prostate cancer cell lines were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). The PC3 prostate cancer cell line was cultured in Roswell Park Memorial Institute 1640 medium (RPMI-1640) and supplemented with 10% (v/v) Fetal Calf serum (FCS) (Thermo Fisher Scientific, Waltham, MA, USA), 50 units/ml penicillin, and 100 μg/mL streptomycin (Thermo Fisher Scientific). Cells tested negative for Mycoplasma.

Hyperinsulinaemic Rag1-/- mouse model treated with unacylated ghrelin (UAG)

To determine the metabolic effect of UAG in an engraftable mouse model of hyperinsulinaemia [69], male recombination-activation gene deficient mice (B6.SVJ129-Rag1tm1Bal/Arc; Rag1-/-) (Jackson Laboratories, supplied by Animal Resource Centre, Murdoch, WA, Australia) were weaned onto a diet of normal chow (chow) or a Western-style, high-fat diet (HFD; 23% fat, SF04-027, Specialty Feeds, Glen Forrest, WA) [69]. After two weeks on the diet, mice were anaesthetised and subcutaneously injected into the left flank with 1×106 PC3 cells diluted 1:1 in growth factor reduced, phenol red-free Matrigel (Corning, Corning, NY, USA). Tumours were allowed to grow until a volume of approximately 50–100 mm3 was reached, when mice were randomly divided into two experimental groups. Mice then received daily intraperitoneal injections of 100 μg/kg UAG (Mimotopes, Mulgrave, VIC, Australia) (n = 6 HFD, n = 10 chow), a dose previously determined to inhibit breast cancer growth in vivo [8], or phosphate buffered saline (PBS) control (n = 8 HFD, n = 10 chow) for 16 days. Tumour volume was calculated by measuring subcutaneous tumour length and width twice weekly using digital calipers (ProSciTech, Kirwan, QLD, Australia). Tumour volume was calculated using the equation ‘tumour volume = (width × length2)/2’ [70]. Body-weight was measured twice weekly.

In order to determine the metabolic effects of the diet, intraperitoneal (i.p.) glucose tolerance tests were performed (n = 7 chow PBS group; n = 6 chow UAG group; n = 5 chow PBS group; n = 6 chow PBS group) as previously described [69]. Briefly, mice were fasted for 16 hours and baseline glucose levels measured in tail-tip blood using OneTouch Ultra Blood Glucose Monitoring System test strips (Accu-Chek Performa, Roche, Basel, Switzerland). Glucose (20% solution, 2 g/kg) was injected i.p. and blood glucose levels assessed 15, 30, 60, and 120 minutes post injection. At experimental endpoint (fourteen days of treatment, or ethical endpoint) mice were euthanised using 70% carbon dioxide followed by cervical dislocation after death was confirmed. Ethical endpoint was based on the tumour volume reaching 1,000 mm3 or a combination of signs of stress including increased heart rate, inactivity, reduced interaction with cage mates, abnormal posture and/or >20% body weight loss. At endpoint, tumours and adipose tissue (epididymal fat pad and interscapular brown adipose tissue) were excised and weighed. Fasting blood glucose was measured at endpoint and blood collected post mortem by cardiac puncture for serum biochemical measurements.

All mice were housed under pathogen-free conditions in individually-ventilated cages, at a room temperature of 20–23 °C, with a 12-hour light-dark cycle. All methods were conducted in accordance with ethical guidelines and regulations and ethics approval from the University of Queensland and Queensland University of Technology Animal Ethics Committees and ethics approval for cell line (LNCaP) use was granted from the Queensland University of Technology Human Research Ethics Committee.

Hormone measurement

Fasting serum insulin and total and acyl-ghrelin levels were determined by ELISA (EMD Merck Millipore Group, Darmstadt, Germany). Absorbance at 450 and 595 nm was determined using a FLUOstar Omega plate reader and software (BMG Labtech, Offenburg, Germany), with absorbance values interpolated using linear regression.

Surrogate indices of insulin resistance, insulin sensitivity, and steady state β-cell function were determined using the homeostatic model for assessment calculator (HOMA2), available from the Oxford Centre for Diabetes, Endocrinology and Metabolism [71], using measured fasting glucose and insulin levels. Unacylated ghrelin (UAG) levels were estimated by subtracting measured serum acyl-ghrelin levels from serum total ghrelin levels.

Histological tissue analysis

Tissue was processed and embedded in paraffin before sectioning (5 μM sections). Immunohistochemistry was performed on tumour sections to investigate the expression of the proliferation marker Ki67 (rabbit anti-Ki67 primary antibody, undiluted, Roche, Basel, Switzerland) and CD31, a marker of angiogenesis (rabbit anti-mouse CD31 primary antibody diluted 1:50 in antibody diluent; Abcam, Cambridge, UK). Sections were counterstained with Mayer’s haematoxylin, dehydrated, and mounted with coverslips using D.P.X neutral mounting medium (Sigma-Aldrich) and observed using an Olympus BX41/702 microscope (U-CMAD3) (Shinjuku, Tokyo Metropolis, Japan).

Statistics

Statistical analyses were performed using GraphPad Prism v.6.01 software (GraphPad Software, San Diego, CA). Kruskal-Wallis (three or more groups) and Mann-Whitney U-test (two groups) tests used for non-normally distributed data, while a two-way ANOVA with Tukey’s post-hoc test used for normally distributed data. P ≤ 0.05 was considered to be statistically significant.

Results

Rag1-/- mice fed a high-fat diet show symptoms of metabolic dysregulation

As reported in our previous study [69], mice fed a Western-style 23% fat diet (HFD) developed symptoms of metabolic syndrome, including hyperinsulinaemia and increased lipid accumulation in the liver and skeletal muscle, compared to normal chow-fed controls. After 7 weeks on the diets, mice fed an HFD developed significantly impaired glucose tolerance 30 (P = 0.011, n = 7, Fig 1A) and 60 minutes (P = 0.019) after glucose challenge compared to control mice fed normal chow. Similarly, body-weight (P = 0.003, Fig 1B) and the weight of white adipose epididymal fat pad deposits (n = 8, P = 0.004, Fig 1C) and interscapular brown adipose tissue (P = 0.0002, n = 8, Fig 1D) were significantly greater at endpoint in HFD-fed mice compared to those fed normal chow. No significant differences in tumour volume or weight (Fig 1F–1H) were observed six weeks after subcutaneous xenograft implantation between mice fed HFD or normal chow. This was expected and mirrors previous results where significant differences in tumour size were observed only after eight weeks [69].

thumbnail
Fig 1. Unacylated ghrelin (UAG) affects glucose tolerance but has no effect on tumour volume or other metabolic parameters.

Rag1-/- mice fed a 23% high-fat diet (HFD) or chow were injected with subcutaneous PC3 xenografts and administered UAG (100μg/kg/day, i.p.) (n = 6 HFD, n = 10 LFD) or PBS control (n = 8 HFD, n = 10 chow) once tumours were palpable. Mean ± s.e.m. * P ≤ 0.05. (A) HFD-fed UAG-treated mice (n = 6) had significantly lower blood glucose 30 min post-glucose challenge compared to HFD-fed PBS treated mice (n = 8), determined by intraperitoneal glucose tolerance test (IPGTT). Mean ± s.e.m. Two-way ANOVA. * P = 0.025. (B) Body weight (g) of mice at endpoint was higher in the HFD-fed mice compared to the chow fed mice (*P<0.05), but was not different between the UAG and PBS groups (P = 0.08). (C) Epididymal fat pad weight (g) was greater in the HFD-PBS group compared to the chow fed-PBS group (*P<0.05), but not the UAG and PBS treatment groups (P = 0.26). (D) Interscapular brown adipose tissue weight (g) was increased in the HFD-PBS group compared to the normal chow groups (P = 0.0002), but not significantly different in UAG-treated mice compared to PBS-treated mice (P = 0.12). Mean ± s.e.m. Mann-Whitney U-test. (E) Fasting blood glucose (mM) was not altered in UAG-treated compared to PBS-treated mice on either diet (P = 0.50). Mean ± s.e.m. (F) Tumour volume (mm3) measured over time (P = 0.57), (G) and tumour volume (mm3) (P = 0.55) and (H) weight (g) at experimental endpoint were not significantly different between UAG- and PBS-treated mice fed HFD or chow. Mean ± s.e.m. Mann-Whitney U-test. (I) Fasting blood insulin (ng/ml) was not altered in UAG-treated compared to PBS-treated mice on either diet (P = 0.90). Mean ± s.e.m. Mann-Whitney U-test. (J) Insulin resistance (HOMA-IR) (P = 0.70), (K) steady state β-cell function (HOMA%B) (P = 0.22) and (L) insulin sensitivity (HOMA%S) (P = 0.70) were not altered in UAG-treated compared to PBS-treated mice or by either diet. (M) Plasma acyl-ghrelin was not altered in mice treated with UAG compared to other mice. (N) Plasma total ghrelin and (O) UAG levels in mice administered UAG (100μg/kg/day) compared to mice treated with PBS. Mean ± s.e.m. Mann-Whitney U-test.

https://doi.org/10.1371/journal.pone.0198495.g001

No effect of intraperitoneal administration of UAG on PC3 xenograft growth in obese, hyperinsulinaemic Rag1-/- mice

No significant differences in tumour volume over the treatment period or tumour weight (P = 0.57) and volume at endpoint (P = 0.55) were observed between UAG- and vehicle control (PBS)-treated obese mice (14 days of treatment) (Mann-Whitney test, Fig 1F–1H). Additionally, no difference in immunohistochemical staining of tumour xenografts for the proliferation marker Ki67 or the angiogenesis marker CD31 was observed between UAG or PBS treated groups, or mice fed normal chow or high-fat diet (HFD) (S1 Fig). Body-weight was reduced in the UAG treatment group fed HFD compared to the PBS treated group, however, the difference was not statistically significant (P = 0.08) (Fig 1B). No metabolic changes were observed in response to UAG treatment (Fig 1B–1E and 1I–1L). While there was a significant difference in fasting blood glucose 30 minutes following glucose challenge in HFD-fed UAG-treated mice compared to PBS controls at endpoint (after 16 days of treatment) (21.6 ± 1.2mM, n = 6 vs 25.4 ± 1.9mM, n = 5, P = 0.02, two-way ANOVA with post-hoc test, Fig 1A), this was not observed at other time points, suggesting that there was no major change in glucose tolerance. There was no significant difference in fasting blood glucose (P = 0.50), blood insulin concentration (P = 0.90, Mann-Whitney U-test, Fig 1I), insulin resistance (P = 0.70, Mann-Whitney U-test, Fig 1J), or insulin sensitivity (P = 0.70, Mann-Whitney U-test, Fig 1L) with UAG treatment compared to PBS control. Plasma acyl-ghrelin (Fig 1M) and total ghrelin levels (Fig 1N) were not altered by diet or UAG treatment, however increased serum UAG levels upon UAG administration was confirmed by ELISA (Fig 1O).

Discussion

It has recently been recognised that UAG can act as a ghrelin inhibitor under some conditions, reducing ghrelin-mediated increases in plasma glucose [22, 26, 28, 72] and lipids [27, 29]. As the ghrelin axis also plays a role in the progression of a number of endocrine-related cancers [5, 3234], including prostate cancer [5, 43], we hypothesised that UAG has beneficial effects in advanced prostate cancer associated with metabolic syndrome. To evaluate this hypothesis we examined the effect of UAG on prostate cancer cell line xenograft growth in vivo.

In our diet-induced hyperinsulinaemic Rag1-/- mouse model [69], we investigated the effect of supraphysiological systemic UAG treatment (100μg/kg/day) on metabolic parameters and PC3 prostate cancer xenograft growth. No differences in metabolic parameters (fasting blood glucose, fasting blood insulin, insulin resistance, steady-state β-cell function, and insulin sensitivity) were observed following UAG treatment in HFD-fed mice. Other studies have found that UAG prevents insulin resistance and hyperglycaemia in short-term HFD-fed mice [73], observations which may stem from the ability of UAG to cross the blood-brain barrier and oppose the central actions of ghrelin on energy homeostasis [74]. Furthermore, in human clinical trials UAG improved glucose and lipid metabolism in healthy [29] and diabetic patients [28]. In our study, a decrease in body-weight, epididymal fat pad weight, and interscapular brown adipose tissue was observed in HFD-fed UAG-treated mice but this difference was not statistically significant. UAG did significantly reduce blood glucose levels at 30 minutes post-glucose challenge in HFD, but not mice on a normal chow diet, however. This is similar to other studies, which only found positive effects of UAG on glucose tolerance in obese patients [72]. Similarly, in clinical trials AZP-531 (a cyclised, truncated analogue of UAG) improved food-related behaviour, waist circumference, and glucose tolerance in Prader-Willi syndrome patients, but had no effect on body weight [31]. AZP-531 also prevents HFD-induced weight gain, insulin resistance, and impairment of glucose tolerance in mice [27].

To the best of our knowledge, this is the first report on the effects of UAG on cancer cell line xenograft growth in vivo. While our study and others show somewhat promising effects of UAG treatment on metabolic parameters, systemic UAG administration had no effect on prostate tumour xenograft size in mice fed a normal chow or high-fat diet. While preliminary, our study suggests that UAG administration, or targeting of endocrine UAG, may have limited therapeutic potential for prostate cancer–in patients with and without symptoms of metabolic syndrome.

Supporting information

S1 Fig. Unacylated ghrelin (UAG) has no effect on tumour histopathology or immunohistochemical markers for proliferation or angiogenesis.

Immunohistochemistry for (A) the proliferation marker Ki67 and (B) the endothelial cell marker CD31, show no difference in positive staining (brown) in PC3 tumour xenografts from mice treated with UAG or PBS in the normal chow or high fat diet (HFD). Arrows show examples of positively stained cells.

https://doi.org/10.1371/journal.pone.0198495.s001

(TIFF)

References

  1. 1. Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, et al. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86(12): 5992. pmid:11739476
  2. 2. Tschöp M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407(6806): 908–13. pmid:11057670
  3. 3. Tschöp M, Wawarta R, Riepl RL, Friedrich S, Bidlingmaier M, Landgraf R, et al. Post-prandial decrease of circulating human ghrelin levels. J. Endocrinol. Invest. 2001;24(6): RC19–21. pmid:11434675
  4. 4. Toshinai K, Mondal MS, Nakazato M, Date Y, Murakami N, Kojima M, et al. Upregulation of Ghrelin expression in the stomach upon fasting, insulin-induced hypoglycemia, and leptin administration. Biochem. Biophys. Res. Commun. 2001;281(5): 1220–5. pmid:11243865
  5. 5. Chopin LK, Seim I, Walpole CM, Herington AC. The ghrelin axis—does it have an appetite for cancer progression? Endocrine Rev. 2012;33(6): 849–91.
  6. 6. Kojima M, Hosoda H, Date Y, Nakazato M, Matsuo H, Kangawa K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature. 1999;402(6762): 656–60. pmid:10604470
  7. 7. Chopin L, Walpole C, Seim I, Cunningham P, Murray R, Whiteside E, et al. Ghrelin and cancer. Mol. Cell. Endocrinol. 2011;340(1): 65–9. pmid:21616120
  8. 8. CheukMan Cherie A, Kara LB, Maria MD, Brid C, Jason EC, John BF, et al. Des-acyl ghrelin suppresses breast cancer cell growth in vitro and in vivo. Meeting Abstracts: Endocrine Society; 2016. p. FRI-065.
  9. 9. Docanto MM, Yang F, Callaghan B, Au CC, Ragavan R, Wang X, et al. Ghrelin and des-acyl ghrelin inhibit aromatase expression and activity in human adipose stromal cells: suppression of cAMP as a possible mechanism. Breast Cancer Res. Treat. 2014;147(1): 193–201. pmid:25056185
  10. 10. Callaghan B, Furness JB. Novel and conventional receptors for ghrelin, desacyl-ghrelin, and pharmacologically related compounds. Pharmacol. Rev. 2014;66(4):984–1001. pmid:25107984
  11. 11. Yang J, Zhao TJ, Goldstein JL, Brown MS. Inhibition of ghrelin O-acyltransferase (GOAT) by octanoylated pentapeptides. Proc. Natl. Acad. Sci. USA 2008;105(31): 10750–5. pmid:18669668
  12. 12. Gutierrez JA, Solenberg PJ, Perkins DR, Willency JA, Knierman MD, Jin Z, et al. Ghrelin octanoylation mediated by an orphan lipid transferase. Proc. Natl. Acad. Sci. USA 2008;105(17): 6320–5. pmid:18443287
  13. 13. Baldanzi G, Filigheddu N, Cutrupi S, Catapano F, Bonissoni S, Fubini A, et al. Ghrelin and des-acyl ghrelin inhibit cell death in cardiomyocytes and endothelial cells through ERK1/2 and PI 3-kinase/AKT. J. Cell Biol. 2002;159(6):1029–37. pmid:12486113
  14. 14. Cassoni P, Ghe C, Marrocco T, Tarabra E, Allia E, Catapano F, et al. Expression of ghrelin and biological activity of specific receptors for ghrelin and des-acyl ghrelin in human prostate neoplasms and related cell lines. Eur. J. Endocrinol. 2004;150(2): 173–84. pmid:14763915
  15. 15. Cassoni P, Papotti M, Ghe C, Catapano F, Sapino A, Graziani A, et al. Identification, characterization, and biological activity of specific receptors for natural (ghrelin) and synthetic growth hormone secretagogues and analogs in human breast carcinomas and cell lines. J. Clin. Endocrinol. Metab. 2001;86(4): 1738–45. pmid:11297611
  16. 16. Gauna C, Delhanty PJD, Hofland LJ, Janssen JAMJL, Broglio F, Ross RJM, et al. Ghrelin stimulates, whereas des-octanoyl ghrelin inhibits, glucose output by primary hepatocytes. J. Clin. Endocrinol. Metab. 2005;90(2): 1055–60. pmid:15536157
  17. 17. Toshinai K, Yamaguchi H, Sun Y, Smith RG, Yamanaka A, Sakurai T, et al. Des-acyl ghrelin induces food intake by a mechanism independent of the growth hormone secretagogue receptor. Endocrinology. 2006;147(5): 2306–14. pmid:16484324
  18. 18. Filigheddu N, Gnocchi VF, Coscia M, Cappelli M, Porporato PE, Taulli R, et al. Ghrelin and des-acyl ghrelin promote differentiation and fusion of C2C12 skeletal muscle cells. Mol. Biol. Cell 2007;18(3): 986–94. pmid:17202410
  19. 19. Delhanty PJ, van der Eerden BC, van der Velde M, Gauna C, Pols HA, Jahr H, et al. Ghrelin and unacylated ghrelin stimulate human osteoblast growth via mitogen-activated protein kinase (MAPK)/phosphoinositide 3-kinase (PI3K) pathways in the absence of GHS-R1a. J. Endocrinol. 2006;188(1): 37–47. pmid:16394173
  20. 20. Delhanty PJ, Sun Y, Visser JA, van Kerkwijk A, Huisman M, van Ijcken WF, et al. Unacylated ghrelin rapidly modulates lipogenic and insulin signaling pathway gene expression in metabolically active tissues of GHSR deleted mice. PloS one. 2010;5(7): e11749. pmid:20668691
  21. 21. Baragli A, Ghe C, Arnoletti E, Granata R, Ghigo E, Muccioli G. Acylated and unacylated ghrelin attenuate isoproterenol-induced lipolysis in isolated rat visceral adipocytes through activation of phosphoinositide 3-kinase gamma and phosphodiesterase 3B. Biochim. Biophys. Acta. 2011;1811(6): 386–96. pmid:21435395
  22. 22. Broglio F, Gottero C, Prodam F, Gauna C, Muccioli G, Papotti M, et al. Non-acylated ghrelin counteracts the metabolic but not the neuroendocrine response to acylated ghrelin in humans. J. Clin. Endocrinol. Metab. 2004;89(6): 3062–5. pmid:15181099
  23. 23. Au CC, Docanto MM, Zahid H, Raffaelli FM, Ferrero RL, Furness JB, et al. Des-acyl ghrelin inhibits the capacity of macrophages to stimulate the expression of aromatase in breast adipose stromal cells. J. Steroid Biochem. Mol. Biol. 2017;170: 49–53. pmid:27423512
  24. 24. Kumar R, Salehi A, Rehfeld JF, Höglund P, Lindström E, Håkanson R. Proghrelin peptides: Desacyl ghrelin is a powerful inhibitor of acylated ghrelin, likely to impair physiological effects of acyl ghrelin but not of obestatin: A study of pancreatic polypeptide secretion from mouse islets. Reg. Peptides. 2010;164(2–3): 65–70. http://doi.org/10.1016/j.regpep.2010.06.005.
  25. 25. Neary NM, Druce MR, Small CJ, Bloom SR. Acylated ghrelin stimulates food intake in the fed and fasted states but desacylated ghrelin has no effect. Gut. 2006;55(1): 135.
  26. 26. Gauna C, Meyler FM, Janssen JAMJL, Delhanty PJD, Abribat T, van Koetsveld P, et al. Administration of acylated ghrelin reduces insulin sensitivity, whereas the combination of acylated plus unacylated ghrelin strongly improves insulin sensitivity. J. Clin. Endocrinol. Metab. 2004;89(10): 5035–42. pmid:15472202
  27. 27. Delhanty PJ, Huisman M, Baldeon-Rojas LY, van den Berge I, Grefhorst A, Abribat T, et al. Des-acyl ghrelin analogs prevent high-fat-diet-induced dysregulation of glucose homeostasis. FASEB J.. 2013;27(4):1690–700. Epub 2013/01/10. pmid:23299855
  28. 28. Ozcan B, Neggers SJ, Miller AR, Yang HC, Lucaites V, Abribat T, et al. Does des-acyl ghrelin improve glycemic control in obese diabetic subjects by decreasing acylated ghrelin levels? Eur. J. Endocrinol. 2014;170(6):799–807. Epub 2013/07/19. pmid:23864339
  29. 29. Benso A S-P D, Prodam F, Gramaglia E, Granata R, van der Lely AJ, Ghigo E, Broglio F. Metabolic effects of overnight continuous infusion of unacylated ghrelin in humans. Eur. J. Endocrinol. 2012;166(5): 911. pmid:22379116
  30. 30. Allas S, Delale T, Ngo N, Julien M, Sahakian P, Ritter J, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of AZP-531, a first-in-class analogue of unacylated ghrelin, in healthy and overweight/obese subjects and subjects with type 2 diabetes. Diabetes Obesity Metab. 2016;18(9): 868–74. pmid:27063928
  31. 31. Allas S, Caixàs A, Poitou C, Coupaye M, Thuilleaux D, Lorenzini F, et al. AZP-531, an unacylated ghrelin analog, improves food-related behavior in patients with Prader-Willi syndrome: A randomized placebo-controlled trial. PloS one. 2018;13(1): e0190849. pmid:29320575
  32. 32. Fung JNT, Jeffery PL, Lee JD, Seim I, Roche D, Obermair A, et al. Silencing of ghrelin receptor expression inhibits endometrial cancer cell growth in vitro and in vivo. Am. J. Physiol. 2013;305(2): E305–13.
  33. 33. Lin TC, Hsiao M. Ghrelin and cancer progression. Biochim. Biophys. Acta. 2017;1868(1): 51–7. pmid:28238732
  34. 34. Grönberg M, Ahlin C, Naeser Y, Janson ET, Holmberg L, Fjällskog M-L. Ghrelin is a prognostic marker and a potential therapeutic target in breast cancer. PloS one. 2017;12(4):e 0176059. pmid:28419141
  35. 35. Zhu J, Yao J, Huang R, Wang Y, Jia M, Huang Y. Ghrelin promotes human non-small cell lung cancer A549 cell proliferation through PI3K/Akt/mTOR/P70S6K and ERK signaling pathways. Biochem. Biophys. Res. Commun. 2018;498(3): 616–20. https://doi.org/10.1016/j.bbrc.2018.03.031. pmid:29524402
  36. 36. American Cancer Society. Cancer Facts & Figures 2018. Atlanta: American Cancer Society; 2018.
  37. 37. Bertaccini A, Pernetti R, Marchiori D, Pagotto U, Palladoro F, Palmieri F, et al. Variations in blood ghrelin levels in prostate cancer patients submitted to hormone suppressive treatment. Anticancer Res. 2009;29(4): 1345–8. pmid:19414385
  38. 38. Mungan NA, Eminferzane S, Mungan AG, Yesilli C, Seckiner I, Can M, et al. Diagnostic value of serum ghrelin levels in prostate cancer. Urol. Int. 2008;80(3):245–8. pmid:18480624
  39. 39. Malendowicz W, Ziolkowska A, Szyszka M, Kwias Z. Elevated blood active ghrelin and unaltered total ghrelin and obestatin concentrations in prostate carcinoma. Urol. Int. 2009;83(4): 471–5. pmid:19996657
  40. 40. Jeffery PL, Herington AC, Chopin LK. Expression and action of the growth hormone releasing peptide ghrelin and its receptor in prostate cancer cell lines. J. Endocrinol. 2002;172(3): R7–R11. pmid:11874717
  41. 41. Yeh AH, Jeffery PL, Duncan RP, Herington AC, Chopin LK. Ghrelin and a novel preproghrelin isoform are highly expressed in prostate cancer and ghrelin activates mitogen-activated protein kinase in prostate cancer. Clin. Cancer Res. 2005;11(23): 8295–303. pmid:16322288
  42. 42. Lanfranco F, Baldi M, Cassoni P, Bosco M, Ghe C, Muccioli G. Ghrelin and prostate cancer. Vitamins Hormones. 2008;77: 301–24. pmid:17983862
  43. 43. Seim I, Jeffery PL, de Amorim L, Walpole CM, Fung J, Whiteside EJ, et al. Ghrelin O-acyltransferase (GOAT) is expressed in prostate cancer tissues and cell lines and expression is differentially regulated in vitro by ghrelin. Repro. Biol. Endocrinol. 2013;11(1): 70.
  44. 44. Duxbury MS, Waseem T, Ito H, Robinson MK, Zinner MJ, Ashley SW, et al. Ghrelin promotes pancreatic adenocarcinoma cellular proliferation and invasiveness. Biochem. Biophys. Res. Commun. 2003;309(2): 464–8. pmid:12951072
  45. 45. Waseem T, Duxbury M, Ito H, Ashley SW, Robinson MK. Exogenous ghrelin modulates release of pro-inflammatory and anti-inflammatory cytokines in LPS-stimulated macrophages through distinct signaling pathways. Surgery. 2008;143(3): 334–42. pmid:18291254
  46. 46. Dixit VD, Weeraratna AT, Yang H, Bertak D, Cooper-Jenkins A, Riggins GJ, et al. Ghrelin and the growth hormone secretagogue receptor constitute a novel autocrine pathway in astrocytoma motility. J. Biol. Chem. 2006;281(24): 16681–90. pmid:16527811
  47. 47. Hammarsten J, Hogstedt B. Hyperinsulinaemia: a prospective risk factor for lethal clinical prostate cancer. Eur. J. Cancer 2005;41(18): 2887–95. pmid:16243513
  48. 48. Ma J, Li H, Giovannucci E, Mucci L, Qiu W, Nguyen PL, et al. Prediagnostic body-mass index, plasma C-peptide concentration, and prostate cancer-specific mortality in men with prostate cancer: a long-term survival analysis. Lancet Oncol. 2008;9(11): 1039–47. pmid:18835745
  49. 49. Steele B, Thomas CC, Henley SJ, Massetti GM, Galuska DA, Agurs-Collins T, et al. Vital Signs: Trends in Incidence of Cancers Associated with Overweight and Obesity—United States, 2005–2014 U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, 2017. Morb. Mort. Wkly Rep. 2017; 66(39):1052–1058.
  50. 50. Souaze F, Dupouy S, Viardot-Foucault V, Bruyneel E, Attoub S, Gespach C, et al. Expression of neurotensin and NT1 receptor in human breast cancer: a potential role in tumor progression. Cancer Res. 2006;66(12): 6243–9. pmid:16778199
  51. 51. Zhang Y, Zhu S, Yi L, Liu Y, Cui H. Neurotensin receptor1 antagonist SR48692 reduces proliferation by inducing apoptosis and cell cycle arrest in melanoma cells. Mol. Cell. Biochem. 2014;389(1–2): 1–8. pmid:24357116
  52. 52. Brown M, Vale W. Effects of neurotensin and substance P on plasma insulin, glucagon and glucose levels. Endocrinology. 1976;98(3): 819–22. pmid:1261503
  53. 53. Sehgal I, Powers S, Huntley B, Powis G, Pittelkow M, Maihle NJ. Neurotensin is an autocrine trophic factor stimulated by androgen withdrawal in human prostate cancer. Proc. Natl. Acad. Sci. USA. 1994;91(11): 4673–7. Epub 1994/05/24. pmid:8197117
  54. 54. Vias M, Burtt G, Culig Z, Veerakumarasivam A, Neal DE, Mills IG. A role for neurotensin in bicalutamide resistant prostate cancer cells. The Prostate. 2007;67(2): 190–202. pmid:17044078
  55. 55. Vidal Samuel J, Rodriguez-Bravo V, Quinn SA, Rodriguez-Barrueco R, Lujambio A, Williams E, et al. A targetable GATA2-IGF2 axis confers aggressiveness in lethal prostate cancer. Cancer Cell. 2015;27(2): 223–39. http://dx.doi.org/10.1016/j.ccell.2014.11.013. pmid:25670080
  56. 56. Perez-Cornago A, Key TJ, Allen NE, Fensom GK, Bradbury KE, Martin RM, et al. Prospective investigation of risk factors for prostate cancer in the UK Biobank cohort study. Br. J. Cancer. 2017;117: 1562. pmid:28910820
  57. 57. Discacciati A, Orsini N, Wolk A. Body mass index and incidence of localized and advanced prostate cancer—a dose-response meta-analysis of prospective studies. Annals Oncol. 2012;23(7):1665–71. pmid:22228452
  58. 58. Gunter JH, Sarkar PL, Lubik AA, Nelson CC. New players for advanced prostate cancer and the rationalisation of insulin-sensitising medication. Int. J. Cell Biol. 2013;2013: 834684. pmid:23573093
  59. 59. World Cancer Research Fund International/American Institute for Cancer Research Continuous Update Project Report: Diet, Nutrition, Physical Activity, and Prostate Cancer. 2014. www.wcrf.org/sites/default/files/Prostate-Cancer-2014-Report.pdf
  60. 60. Grayhack JT, Keeler TC, Kozlowski JM. Carcinoma of the prostate. Hormonal therapy. Cancer. 1987;60(3 Suppl): 589–601. pmid:3297288
  61. 61. Vela I, Gregory LS, Gardiner EM, Clements JA, Nicol DL. Bone and prostate cancer cell interactions in metastatic prostate cancer. BJU International. 2007;99(4): 735–42. pmid:17166237
  62. 62. Loberg RD, Gayed BA, Olson KB, Pienta KJ. A paradigm for the treatment of prostate cancer bone metastases based on an understanding of tumor cell-microenvironment interactions. J. Cell Biol. 2005;96(3): 439–46. pmid:15988761
  63. 63. Sturge J, Caley MP, Waxman J. Bone metastasis in prostate cancer: emerging therapeutic strategies. Nature Rev. Clin. Oncol. 2011;8:357–68. pmid:21556025
  64. 64. Senmaru T, Fukui M, Okada H, Mineoka Y, Yamazaki M, Tsujikawa M, et al. Testosterone deficiency induces markedly decreased serum triglycerides, increased small dense LDL, and hepatic steatosis mediated by dysregulation of lipid assembly and secretion in mice fed a high-fat diet. Metab. Clin. Exp. 2013;62(6): 851–60. pmid:23332447
  65. 65. Basaria S, Muller DC, Carducci MA, Egan J, Dobs AS. Hyperglycemia and insulin resistance in men with prostate carcinoma who receive androgen-deprivation therapy. Cancer. 2006;106(3): 581–8. pmid:16388523
  66. 66. Lubik AA, Gunter JH, Hendy SC, Locke JA, Adomat HH, Thompson V, et al. Insulin increases de novo steroidogenesis in prostate cancer cells. Cancer Res. 2011;71(17): 5754–64. pmid:21747118
  67. 67. Hsing AW, Sakoda LC, Chua S Jr. Obesity, metabolic syndrome, and prostate cancer. Am. J. Clin. Nut. 2007;86(3): s843–57.
  68. 68. Grossmann M, Wittert G. Androgens, diabetes and prostate cancer. Endocrine Related Cancer. 2012;19(5): F47–62. pmid:22514110
  69. 69. Maugham ML, Thomas PB, Crisp GJ, Philp LK, Shah ET, Herington AC, et al. Insights from engraftable immunodeficient mouse models of hyperinsulinaemia. Sci. Rep. 2017;7(1): 491. pmid:28352127
  70. 70. Scott KL, Kabbarah O, Liang M-C, Ivanova E, Anagnostou V, et al. GOLPH3 modulates mTOR signalling and rapamycin sensitivity in cancer. Nature. 2009; 459: 1085–1090. pmid:19553991
  71. 71. Diabetes Trial Unit. The Oxford Centre for Diabetes EaM. HOMA Calculator University of Oxford. https://www.dtu.ox.ac.uk/homacalculator/index.php.
  72. 72. Tong J, Davis HW, Summer S, Benoit SC, Haque A, Bidlingmaier M, et al. Acute administration of unacylated ghrelin has no effect on basal or stimulated insulin secretion in healthy humans. Diabetes. 2014;63(7): 2309–19. pmid:24550190
  73. 73. Gortan Cappellari G, Zanetti M, Semolic A, Vinci P, Ruozi G, Falcione A, et al. Unacylated ghrelin reduces skeletal muscle reactive oxygen species generation and inflammation and prevents high-fat diet-induced hyperglycemia and whole-body insulin resistance in rodents. Diabetes. 2016;65(4):874–86. pmid:26822085
  74. 74. Banks WA, Tschop M, Robinson SM, Heiman ML. Extent and direction of ghrelin transport across the blood-brain barrier is determined by its unique primary structure. J. Pharmacol. Exper. Therap. 2002;302(2): 822–7. pmid:12130749