1
|
Murata D, Azuma K, Matsuo N, Murotani K, Matama G, Kawahara A, Sasada T, Tokito T, Hoshino T. Survival and biomarkers for cachexia in non-small cell lung cancer receiving immune checkpoint inhibitors. Cancer Med 2023; 12:19471-19479. [PMID: 37712645 PMCID: PMC10587946 DOI: 10.1002/cam4.6549] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/16/2023] Open
Abstract
BACKGROUND The presence of cachexia negatively impacts the prognosis of patients with cancer. However, the mechanisms behind the development of cachexia and its prognostic impact on immunotherapy efficacy are not fully understood. MATERIALS AND METHODS We retrospectively screened patients with advanced or recurrent non-small cell lung cancer (NSCLC) who received PD-1/PD-L1 inhibitor monotherapy. Among 183 patients, pre-treatment plasma samples were available from 100 patients. We defined cancer cachexia as weight loss of at least 5% during the past 6 months or weight loss of at least 2% and BMI <20. We analyzed 75 soluble immune mediators in pre-treatment plasma samples to explore the possible mechanisms behind the development of cancer cachexia. We also investigated whether cancer cachexia affects prognosis. RESULTS Among 100 patients, 35 had cancer cachexia. Logistic regression analysis identified ghrelin, c-reactive protein (CRP), pentraxin-3 (PTX-3), and osteopontin (OPN) as factors associated with cachexia. Patients with cachexia had worse progression-free survival (PFS) and overall survival (OS), although we did not detect statistically significant differences. Analyzing the soluble immune mediators associated with cachexia, the combination of cachexia and PTX-3 or OPN expression levels was predictive for PFS and the combination of cachexia and CRP or OPN expression levels was predictive for OS. CONCLUSIONS Pre-treatment ghrelin, CRP, PTX-3, and OPN may be associated with cachexia. Among patients with NSCLC who received PD-1/L1 inhibitor monotherapy, those with cachexia had worse survival than those without cachexia. Larger studies will be required to confirm our data and better understand the mechanisms behind the development of cachexia.
Collapse
Affiliation(s)
- Daiki Murata
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| | - Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| | - Norikazu Matsuo
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| | - Kenta Murotani
- Biostatistics CenterKurume University School of MedicineFukuokaJapan
| | - Goushi Matama
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| | - Akihiko Kawahara
- Department of Diagnostic PathologyKurume University HospitalFukuokaJapan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center and Division of Cancer ImmunotherapyKanagawa Cancer Center Research InstituteKanagawaJapan
| | - Takaaki Tokito
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal MedicineKurume University School of MedicineFukuokaJapan
| |
Collapse
|
2
|
Stojsavljevic-Shapeski S, Virovic-Jukic L, Tomas D, Duvnjak M, Tomasic V, Hrabar D, Kralj D, Budimir I, Barsic N, Ljubicic N. Expression of adipokine ghrelin and ghrelin receptor in human colorectal adenoma and correlation with the grade of dysplasia. World J Gastrointest Surg 2021; 13:1708-1720. [PMID: 35070075 PMCID: PMC8727187 DOI: 10.4240/wjgs.v13.i12.1708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/20/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Ghrelin is an adipokine that plays an important role in energy balance. Expression of ghrelin and ghrelin receptor has been investigated in different tissues and tumors. Studies regarding expression of ghrelin and ghrelin receptor in colorectal tumors are scarce and no data on expression of ghrelin and its receptor in colorectal adenomas has been published. Ghrelin and ghrelin receptor were highly expressed in colon carcinoma cells while expression was decreased in less differentiated tumors, presuming that ghrelin might be important in early phases of tumorigenesis.
AIM To investigate the expression of ghrelin and ghrelin receptor in human colorectal adenomas and adjacent colorectal tissue.
METHODS In this prospective study (conducted from June 2015 until May 2019) we included 92 patients (64 male and 28 female) who underwent polypectomy for colorectal adenomas in the Department of Gastroenterology and Hepatology, “Sestre milosrdnice” Clinical Hospital Center in Zagreb, Croatia. After endoscopic removal of colorectal adenoma, an additional sample of colon mucosa in the proximity of the adenoma was collected for pathohistological analysis. Adenomas were graded according to the stage of dysplasia, and ghrelin and ghrelin receptor expression were determined immunohistochemically in both adenoma and adjacent colon tissue using the polyclonal antibody for ghrelin (ab150514, ABCAM Inc, Cambridge, United States) and ghrelin receptor (ab48285, ABCAM Inc, Cambridge, United States). Categorical and nominal variables were described through frequencies and proportions and the difference between specific groups were analyzed with Fisher’s and Fisher-Freeman-Halton’s method respectively. Spearman's rank correlation coefficient was determined for correlation of expression of ghrelin and ghrelin receptor in adenoma and adjacent colon tissue with the grade of adenoma dysplasia.
RESULTS Among 92 patients with colorectal adenoma 43 had adenomas with high-grade dysplasia (46.7%). High expression of ghrelin was 7 times more common in high-grade adenoma compared to low-grade adenomas (13.95% to 2.04%, P = 0.048), while the expression of ghrelin in adjacent colon tissue was low. We found no correlation between ghrelin receptor expression in adenoma and adjacent colon tissue and the grade of colorectal adenoma dysplasia. The most significant correlation was found between ghrelin and ghrelin receptor expression in adenomas with high-grade dysplasia (rho = 0.519, P < 0.001).
CONCLUSION Ghrelin and ghrelin receptor are expressed in colorectal adenoma and adjacent tissue with ghrelin expression being more pronounced in high grade dysplasia as a possible consequence of increased local synthesis.
Collapse
Affiliation(s)
- Sanja Stojsavljevic-Shapeski
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
| | - Lucija Virovic-Jukic
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Davor Tomas
- Ljudevit Jurak Department of Pathology, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
- Department of Pathology, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Marko Duvnjak
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Vedran Tomasic
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
| | - Davor Hrabar
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| | - Dominik Kralj
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
| | - Ivan Budimir
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
| | - Neven Barsic
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
| | - Neven Ljubicic
- Division of Gastroenterology, Department of Internal Medicine, «Sestre Milosrdnice» University Hospital Center, Zagreb 10000, Croatia
- Department of Internal Medicine, School of Medicine, University of Zagreb, Zagreb 10000, Croatia
| |
Collapse
|
3
|
Tegshee B, Kondo K, Soejima S, Muguruma K, Tsuboi M, Kajiura K, Kawakami Y, Kawakita N, Toba H, Yoshida M, Takizawa H, Tangoku A. GHSR methylation-dependent expression of a variant ligand and receptor of the ghrelin system induces thymoma tumorigenesis. Oncol Lett 2021; 22:793. [PMID: 34630704 PMCID: PMC8477069 DOI: 10.3892/ol.2021.13054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 08/19/2021] [Indexed: 12/17/2022] Open
Abstract
Our previous study reported that the DNA methylation of growth hormone secretagogue receptor (GHSR) was significantly higher in thymoma or thymic carcinoma (TC) than in normal thymic tissue samples. Thymic epithelial tumors (TETs) with higher GHSR DNA methylation were associated with significantly worse prognosis than those with lower levels of DNA methylation. Diversified components of the ghrelin-GHSR axis may exert opposing effects in cancer progression, depending on the cancer type in question. However, the precise function of the axis remains unclear. In the present study, the mRNA expression of five key components of the ghrelin system [native ligand ghrelin, variant ligand In-1 ghrelin, native receptor GHSR1a, variant receptor GHSR1b and acylation enzyme ghrelin O-acyltransferase (GOAT)] were examined in 58 TET samples by reverse transcription-quantitative PCR, and protein expression of GHSR1a and GHSR1b was assessed in 20 TETs using immunohistochemistry. The results revealed that In-1 ghrelin, GHSR1b (variant forms) and GOAT were more strongly expressed in thymoma compared with thymic-adjacent tissue. By contrast, no significant differences were observed in the expression of ghrelin and GHSR1a (native forms) between thymoma and thymic tissue. The mRNA expression of In-1 ghrelin and GHSR1b (variant forms) was positively associated with GHSR methylation in thymoma tissue samples. However, a relationship was not found between ghrelin, GHSR1a or GOAT expression (native forms) and GHSR methylation in thymoma. Immunohistochemical analysis revealed that mRNA expression of GHSR1a and GHSR1b generally correlated with expression of the corresponding protein, and that the expression of GHSR1b was increased in advanced-stage TETs. These results indicate that the DNA methylation of GHSR is associated with a shift from native expression (ghrelin and GHSR1a) to variant expression (In-1 ghrelin and GHSR1b), which induces the tumorigenesis of thymoma, but not TC.
Collapse
Affiliation(s)
- Bilguun Tegshee
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8509, Japan
| | - Kazuya Kondo
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8509, Japan
| | - Shiho Soejima
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8509, Japan
| | - Kyoka Muguruma
- Department of Oncological Medical Services, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8509, Japan
| | - Mitsuhiro Tsuboi
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Koichiro Kajiura
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Yukikiyo Kawakami
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Naoya Kawakita
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hiroaki Toba
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Mitsuteru Yoshida
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Hiromitsu Takizawa
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| | - Akira Tangoku
- Department of Thoracic, Endocrine Surgery and Oncology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto-cho, Tokushima 770-8503, Japan
| |
Collapse
|
4
|
Protective and Healing Effects of Ghrelin and Risk of Cancer in the Digestive System. Int J Mol Sci 2021; 22:ijms221910571. [PMID: 34638910 PMCID: PMC8509076 DOI: 10.3390/ijms221910571] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 01/19/2023] Open
Abstract
Ghrelin is an endogenous ligand for the ghrelin receptor, previously known as the growth hormone secretagogue receptor. This hormone is mainly produced by endocrine cells present in the gastric mucosa. The ghrelin-producing cells are also present in other organs of the body, mainly in the digestive system, but in much smaller amount. Ghrelin exhibits a broad spectrum of physiological effects, such as stimulation of growth hormone secretion, gastric secretion, gastrointestinal motility, and food intake, as well as regulation of glucose homeostasis and bone formation, and inhibition of inflammatory processes. This review summarizes the recent findings concerning animal and human data showing protective and therapeutic effects of ghrelin in the gut, and also presents the role of growth hormone and insulin-like growth factor-1 in these effects. In addition, the current data on the possible influence of ghrelin on the carcinogenesis, its importance in predicting the risk of developing gastrointestinal malignances, as well as the potential usefulness of ghrelin in the treatment of cancer, have been presented.
Collapse
|
5
|
Qiu X, Han NS, Yao JX, Yu FR, Lin YY, Zhuang X. Acupuncture Reduced the Risk for Insomnia in Stroke Patients: A Propensity-Score Matched Cohort Study. Front Aging Neurosci 2021; 13:698988. [PMID: 34483881 PMCID: PMC8414891 DOI: 10.3389/fnagi.2021.698988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/20/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Post-stroke insomnia (PSI) affects the quality of life for stroke patients, reduces the likelihood of successful rehabilitation, and produces additional complications following stroke. Previous reports have provided some information regarding PSI risk factors, but little is known concerning protective factors for PSI. This study analyzed the relationship between acupuncture and insomnia in stroke patients and explored the use of acupuncture as a preventive treatment. Methods: Patients diagnosed with stroke from 2010 to 2019 were identified in the case database of the First Affiliated Hospital of Guangzhou University of Chinese These patients followed until 2020, and numerous factors were examined, including gender, age, stroke type, stroke location, and baseline comorbidities. A 1:1 propensity score was used to match an equal number of patients receiving acupuncture with stroke patients who did not receive acupuncture (N = 1,680 for each group). The purpose of the study was to compare the incidence of insomnia in these two stroke cohorts. We used the Cox regression model and Kaplan-Meier method to estimate the risk of insomnia as the outcome event. Results: Compared with the non-acupuncture cohort in general, stroke patients who received acupuncture treatment exhibited a lower risk of insomnia after adjusting for age, gender, stroke type, stroke location, and comorbidities (adjusted hazard ratio HR = 0.27, 95% confidential interval = 0.23 to 0.32). Acupuncture also reduced the risk of PSI for both genders. The respective risks were HR = 0.28 (adjusted) for males and HR = 0.26 (adjusted) for females. Acupuncture also lowered the risk for PSI for different age groups. The risks were HR = 0.22 (adjusted) for individuals 18 to 39 years of age, HR = 0.31 (adjusted) for individuals 40 to 59 years of age, HR = 0.28 (adjusted) for those 60 to 79 years of age, and HR = 0.18 (adjusted) for individuals 80 years of age and older. Concerning the stroke type, regardless of whether the stroke was ischemic, hemorrhagic, or a combination of the two stroke types, patients who received acupuncture exhibited lower risk (adjusted HR = 0.28, 0.17, and 0.49, respectively). Concerning stroke location, except for the cerebral hemispheres (adjusted HR = 1.10, 95% confidential interval = 0.12 to 1.01), the risk of PSI after receiving acupuncture was lower for the frontal lobe (adjusted HR = 0.42), the basal ganglia (adjusted HR = 0.22), the radiation crown (adjusted HR = 0.42), the diencephalon (adjusted HR = 0.20), or multiple partial strokes (adjusted HR = 0.26), the risk of PSI after receiving acupuncture was lower. For all baseline complications, acupuncture reduced the risk of insomnia. The cumulative incidence of insomnia in the acupuncture cohort was significantly lower than the non-acupuncture cohort (log-rank test, P = 0.000). Limitations: First, our research only included patients from a single center. Second, we did not classify the post-stroke insomnia severity. Second, the information was extracted manually. Overall, the sample size was small, and we needed to increase the sample size to strengthen the conclusions. Conclusion: Acupuncture treatment reduced the risk of insomnia in stroke patients. Future research be conducted with increased sample sizes and further elaboration on the specific acupuncture protocols that were used.
Collapse
Affiliation(s)
- Xuan Qiu
- Acupuncture and Rehabilitation Clinical School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Nan Sheng Han
- Acupuncture and Rehabilitation Clinical School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Jie Xiao Yao
- Acupuncture and Rehabilitation Clinical School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Fang Rui Yu
- Acupuncture and Rehabilitation Clinical School, Guangzhou University of Traditional Chinese Medicine, Guangzhou, China
| | - Yan Yan Lin
- Department of Traditional Chinese Medicine, The Second Clinical Medical College, Guangdong Medical University, Dongguan, China
| | - Xun Zhuang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
6
|
Spiridon IA, Ciobanu DGA, Giușcă SE, Căruntu ID. Ghrelin and its role in gastrointestinal tract tumors (Review). Mol Med Rep 2021; 24:663. [PMID: 34296307 PMCID: PMC8335721 DOI: 10.3892/mmr.2021.12302] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Ghrelin, an orexigenic hormone, is a peptide that binds to the growth hormone secretagogue receptor; it is secreted mainly by enteroendocrine cells in the oxyntic glands of the stomach. Ghrelin serves a role in both local and systemic physiological processes, and is implicated in various pathologies, including neoplasia, with tissue expression in several types of malignancies in both in vitro and in vivo studies. However, the precise implications of the ghrelin axis in metastasis, invasion and cancer progression regulation has yet to be established. In the case of gastrointestinal (GI) tract malignancies, ghrelin has shown potential to become a prognostic factor or even a therapeutic target, although data in the literature are inconsistent and unsystematic, with reports untailored to a specific histological subtype of cancer or a particular localization. The evaluation of immunohistochemical expression shows a limited outlook owing to the low number of cases analyzed, and in vivo analyses have conflicting data regarding differences in ghrelin serum levels in patients with cancer. The aim of this review was to examine the relationship between ghrelin and GI tract malignancies to demonstrate the inconsistencies in current results and to highlight its clinical significance in the outcome of these patients.
Collapse
Affiliation(s)
- Irene Alexandra Spiridon
- Department of Pathology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| | | | - Simona Eliza Giușcă
- Department of Pathology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| | - Irina Draga Căruntu
- Department of Histology, 'Grigore T. Popa' University of Medicine and Pharmacy, Iași 700115, Romania
| |
Collapse
|
7
|
Stoyanova I, Lutz D. Ghrelin-Mediated Regeneration and Plasticity After Nervous System Injury. Front Cell Dev Biol 2021; 9:595914. [PMID: 33869167 PMCID: PMC8046019 DOI: 10.3389/fcell.2021.595914] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/24/2021] [Indexed: 12/17/2022] Open
Abstract
The nervous system is highly vulnerable to different factors which may cause injury followed by an acute or chronic neurodegeneration. Injury involves a loss of extracellular matrix integrity, neuronal circuitry disintegration, and impairment of synaptic activity and plasticity. Application of pleiotropic molecules initiating extracellular matrix reorganization and stimulating neuronal plasticity could prevent propagation of the degeneration into the tissue surrounding the injury. To find an omnipotent therapeutic molecule, however, seems to be a fairly ambitious task, given the complex demands of the regenerating nervous system that need to be fulfilled. Among the vast number of candidates examined so far, the neuropeptide and hormone ghrelin holds within a very promising therapeutic potential with its ability to cross the blood-brain barrier, to balance metabolic processes, and to stimulate neurorepair and neuroactivity. Compared with its well-established systemic effects in treatment of metabolism-related disorders, the therapeutic potential of ghrelin on neuroregeneration upon injury has received lesser appreciation though. Here, we discuss emerging concepts of ghrelin as an omnipotent player unleashing developmentally related molecular cues and morphogenic cascades, which could attenuate and/or counteract acute and chronic neurodegeneration.
Collapse
Affiliation(s)
- Irina Stoyanova
- Department of Anatomy and Cell Biology, Medical University Varna, Varna, Bulgaria
| | - David Lutz
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
8
|
Perego S, Sansoni V, Ziemann E, Lombardi G. Another Weapon against Cancer and Metastasis: Physical-Activity-Dependent Effects on Adiposity and Adipokines. Int J Mol Sci 2021; 22:ijms22042005. [PMID: 33670492 PMCID: PMC7922129 DOI: 10.3390/ijms22042005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022] Open
Abstract
Physically active behavior has been associated with a reduced risk of developing certain types of cancer and improved psychological conditions for patients by reducing anxiety and depression, in turn improving the quality of life of cancer patients. On the other hand, the correlations between inactivity, sedentary behavior, and overweight and obesity with the risk of development and progression of various cancers are well studied, mainly in middle-aged and elderly subjects. In this article, we have revised the evidence on the effects of physical activity on the expression and release of the adipose-tissue-derived mediators of low-grade chronic inflammation, i.e., adipokines, as well as the adipokine-mediated impacts of physical activity on tumor development, growth, and metastasis. Importantly, exercise training may be effective in mitigating the side effects related to anti-cancer treatment, thereby underlining the importance of encouraging cancer patients to engage in moderate-intensity activities. However, the strong need to customize and adapt exercises to a patient’s abilities is apparent. Besides the preventive effects of physically active behavior against the adipokine-stimulated cancer risk, it remains poorly understood how physical activity, through its actions as an adipokine, can actually influence the onset and development of metastases.
Collapse
Affiliation(s)
- Silvia Perego
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
| | - Veronica Sansoni
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
- Correspondence: ; Tel.: +39-0266214068
| | - Ewa Ziemann
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, 61-871 Poznań, Poland; or
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, Milano, IRCCS Istituto Ortopedico Galeazzi, 20161 Milan, Italy; (S.P.); or
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, 61-871 Poznań, Poland; or
| |
Collapse
|
9
|
Abstract
Eating food is one of the most complicated behaviours in mammals, especially humans. The primary function of ghrelin is regulation of the appetite level and its stimulation. It is also responsible for the body's energy balance and glucose homeostasis. Ghrelin has been shown to affect many brain structures, which confirms the presence of ghrelin receptors in the brain. Studies are also conducted to assess the possible role of ghrelin in anxiety states and in memory disorders and motor dysfunctions. Ghrelin has been found in saliva and salivary glands, teeth and gums, and in the taste buds of the tongue epithelium; it is also secreted by mucosal cells and gingival fibroblasts. The presence of ghrelin in developmental enamel, especially in odontoblasts and ameloblasts, may suggest its regulatory role in the development of teeth. Patients with chronic periodontitis have significantly higher concentrations of ghrelin in the peripheral blood serum, as compared to the control group. Ghrelin plays a special role in the proliferation of cancer cells and in the development of neoplastic metastases. The abundant presence of ghrelin receptors in cancer cells is considered an important target in the treatment of neoplasms. Ghrelin is a hormone whose multidirectional mechanism of action has not yet been fully understood. However, its ubiquitous occurrence in the human body and its very diverse participation in metabolic processes may prove to be a significant obstacle in achieving the expected clinical effect of ghrelin as an effective drug in selected disease units.
Collapse
|
10
|
Au CC, Furness JB, Britt K, Oshchepkova S, Ladumor H, Soo KY, Callaghan B, Gerard C, Inghirami G, Mittal V, Wang Y, Huang XY, Spector JA, Andreopoulou E, Zumbo P, Betel D, Dow L, Brown KA. Three-dimensional growth of breast cancer cells potentiates the anti-tumor effects of unacylated ghrelin and AZP-531. eLife 2020; 9:56913. [PMID: 32667883 PMCID: PMC7363447 DOI: 10.7554/elife.56913] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/25/2020] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is the most common type of cancer in women and notwithstanding important therapeutic advances, remains the second leading cause of cancer-related death. Despite extensive research relating to the hormone ghrelin, responsible for the stimulation of growth hormone release and appetite, little is known of the effects of its unacylated form, especially in cancer. The present study aimed to characterize effects of unacylated ghrelin on breast cancer cells, define its mechanism of action, and explore the therapeutic potential of unacylated ghrelin or analog AZP-531. We report potent anti-tumor effects of unacylated ghrelin, dependent on cells being cultured in 3D in a biologically-relevant extracellular matrix. The mechanism of unacylated ghrelin-mediated growth inhibition involves activation of Gαi and suppression of MAPK signaling. AZP-531 also suppresses the growth of breast cancer cells in vitro and in xenografts, and may be a novel approach for the safe and effective treatment of breast cancer.
Collapse
Affiliation(s)
- CheukMan C Au
- Department of Medicine, Weill Cornell Medicine, New York, United States.,Centre for Cancer Research, Hudson Institute for Medical Research, Clayton, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Kara Britt
- Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Sofya Oshchepkova
- Department of Medicine, Weill Cornell Medicine, New York, United States
| | - Heta Ladumor
- Department of Medicine, Weill Cornell Medicine, New York, United States.,Weill Cornell Medicine - Qatar, Doha, Qatar
| | - Kai Ying Soo
- Centre for Cancer Research, Hudson Institute for Medical Research, Clayton, Australia
| | - Brid Callaghan
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Australia
| | - Celine Gerard
- Centre for Cancer Research, Hudson Institute for Medical Research, Clayton, Australia
| | - Giorgio Inghirami
- Department of Pathology, Weill Cornell Medical College, New York, United States
| | - Vivek Mittal
- Department of Cardiothoracic Surgery, Department of Cell and Developmental Biology, Neuberger Berman Lung Cancer Center, Weill Cornell Medicine, New York, United States
| | - Yufeng Wang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, United States
| | - Xin Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, United States
| | - Jason A Spector
- Department of Surgery, Weill Cornell Medicine, New York, United States
| | | | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, United States.,Applied Bioinformatics Core, Weill Cornell Medical College, New York, United States
| | - Doron Betel
- Department of Medicine, Weill Cornell Medicine, New York, United States.,Institute for Computational Biomedicine, Weill Cornell Medical College, New York, United States
| | - Lukas Dow
- Department of Medicine, Weill Cornell Medicine, New York, United States
| | - Kristy A Brown
- Department of Medicine, Weill Cornell Medicine, New York, United States.,Centre for Cancer Research, Hudson Institute for Medical Research, Clayton, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, Australia
| |
Collapse
|
11
|
Zhang J, Xie T. Ghrelin inhibits cisplatin-induced MDA-MB-231 breast cancer cell apoptosis via PI3K/Akt/mTOR signaling. Exp Ther Med 2019; 19:1633-1640. [PMID: 32104214 PMCID: PMC7027091 DOI: 10.3892/etm.2019.8398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 02/21/2019] [Indexed: 02/07/2023] Open
Abstract
Ghrelin is a multi-functional peptide, its role on cancer cell apoptosis remains controversial. The present study examined the effects and mechanisms of ghrelin on cisplatin-induced apoptosis in human breast cancer cells. It was identified that ghrelin inhibited apoptosis in MDA-MB-231 cells in vitro and reversed the expression of B-cell lymphoma 2 (Bcl2) and Bcl2-associated X, and cleaved caspase-3 induced by cisplatin. Furthermore, ghrelin activated the phosphoinositide 3-kinases/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway after cisplatin treatment. The effects of ghrelin on the cisplatin-induced apoptosis and PI3K/Akt/mTOR signaling were reversed by the growth hormone secretagogue receptor small interfering RNA. The present study suggests that ghrelin may serve as a novel target for cisplatin resistance and a potential indicator of cisplatin sensitivity in breast cancer treatment.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Oncology, Medical College of Heibei University, Baoding, Hebei 071000, P.R. China
| | - Tianhao Xie
- Department of General Surgery, Affiliated Hospital of Hebei University, Baoding, Hebei 071000, P.R. China
| |
Collapse
|
12
|
Ghrelin Aggravates Prostate Enlargement in Rats with Testosterone-Induced Benign Prostatic Hyperplasia, Stromal Cell Proliferation, and Smooth Muscle Contraction in Human Prostate Tissues. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4748312. [PMID: 31885795 PMCID: PMC6893282 DOI: 10.1155/2019/4748312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/01/2019] [Accepted: 10/30/2019] [Indexed: 12/15/2022]
Abstract
Epidemiologic studies revealed a context between lower urinary tract symptoms (LUTS) suggestive of benign prostatic hyperplasia (BPH) and metabolic syndrome. However, molecular mechanisms underlying this relationship are largely unknown. Prostate enlargement and increased prostate smooth muscle tone are important factors in the pathophysiology of LUTS suggestive of BPH. In the present study, we studied effects of the metabolic hormone ghrelin on prostate enlargement in rats with experimentally induced BPH, growth of cultured stromal cells from human prostate (WPMY-1), and smooth muscle contraction of human prostate tissues. Ghrelin (20 nmol/kg daily, p.o., 2 weeks) increased prostate size in rats with testosterone-induced BPH. Microarray identified 114 ghrelin-upregulated genes (2-fold or more) in these prostates, with possible roles in growth, smooth muscle contraction, or metabolism. 12 genes were selected for further analyses. In human prostate tissues, mRNA levels of 11 of them correlated positively with ghrelin receptor (GHSR) expression, but only two with the degree of BPH. Accordingly, no correlation was evident between GHSR expression level and BPH in human prostate tissues. In WPMY-1 cells, the GHRS agonist MK0677 upregulated 11 of the selected genes. MK0677 induced proliferation of WPMY-1 cells, shown by EdU assay, colony formation, proliferation markers, flow cytometry, and viability. In myographic measurements, GHSR agonists enhanced contractions of human prostate strips. Together, ghrelin may aggravate prostate enlargement, stromal cell growth, and prostate smooth muscle contraction in BPH. Ghrelin may deteriorate urethral obstruction independently from BPH, qualifying the ghrelin system as an attractive new target to be tested for LUTS treatment in BPH.
Collapse
|
13
|
Soleyman-Jahi S, Sadeghi F, Pastaki Khoshbin A, Khani L, Roosta V, Zendehdel K. Attribution of Ghrelin to Cancer; Attempts to Unravel an Apparent Controversy. Front Oncol 2019; 9:1014. [PMID: 31681567 PMCID: PMC6805778 DOI: 10.3389/fonc.2019.01014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022] Open
Abstract
Ghrelin is an endogenous peptide hormone mainly produced in the stomach. It has been known to regulate energy homeostasis, stimulate secretion of growth hormone, and mediate many other physiologic effects. Various effects attributed to ghrelin contribute to many aspects of cancer development and progression. Accordingly, a large body of evidence has emerged about the association of ghrelin with several types of cancer in scales of cell-line, animal, and human studies. However, existing data are controversial. This controversy occurs in two main domains: one is the controversial results in local effects of ghrelin on different types of human cancer cell-lines; the second is the apparent disagreement in the results of in-vitro and clinical studies that investigated ghrelin association to one type of cancer. These inconsistencies have hampered the indications to consider ghrelin as a potential tumor biomarker or therapeutic agent in cancer patients. Previous studies have reviewed different parts of current literature about the ghrelin-cancer relationship. Although they have highlighted these controversial results in various ways, no specific recommendations have been given to address it. In this study, we comprehensively reviewed in-vitro, in-vivo, and clinical studies and attempted to use the following approaches to unravel the inconsistencies detected: (a) to distinguish local and systemic effects of ghrelin in interpreting its summary clinical role in each cancer; (b) scrutinizing factors that regulate local effects of ghrelin and could justify different effects of ghrelin on different cancer cell-lines. These approaches could have notable implications for future in-vitro and clinical studies.
Collapse
Affiliation(s)
- Saeed Soleyman-Jahi
- Division of Gastroenterology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States.,Cancer Immunology Project, Universal Scientific Education and Research Network, St. Louis, MO, United States.,Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadeghi
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project, Universal Scientific Education and Research Network, Tehran, Iran.,Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Pastaki Khoshbin
- Cancer Immunology Project, Universal Scientific Education and Research Network, Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Khani
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Venus Roosta
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kazem Zendehdel
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Maugham ML, Seim I, Thomas PB, Crisp GJ, Shah ET, Herington AC, Brown KA, Gregory LS, Nelson CC, Jeffery PL, Chopin LK. No effect of unacylated ghrelin administration on subcutaneous PC3 xenograft growth or metabolic parameters in a Rag1-/- mouse model of metabolic dysfunction. PLoS One 2018; 13:e0198495. [PMID: 30458004 PMCID: PMC6245673 DOI: 10.1371/journal.pone.0198495] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 11/02/2018] [Indexed: 12/12/2022] Open
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.
Collapse
Affiliation(s)
- Michelle L. Maugham
- 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
- 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
- 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
- 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
- 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
- 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
- Department of Medicine, Weill Cornell Medicine, New York City, New York, United States of America
| | - Laura S. Gregory
- Skeletal Biology and Forensic Anthropology Research Laboratory, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Colleen C. Nelson
- 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
- 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
- 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
| |
Collapse
|
15
|
Herrera-Martínez AD, Gahete MD, Sánchez-Sánchez R, Alors-Perez E, Pedraza-Arevalo S, Serrano-Blanch R, Martínez-Fuentes AJ, Gálvez-Moreno MA, Castaño JP, Luque RM. Ghrelin-O-Acyltransferase (GOAT) Enzyme as a Novel Potential Biomarker in Gastroenteropancreatic Neuroendocrine Tumors. Clin Transl Gastroenterol 2018; 9:196. [PMID: 30297816 PMCID: PMC6175927 DOI: 10.1038/s41424-018-0058-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 08/16/2018] [Accepted: 08/17/2018] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The association between the presence and alterations of the components of the ghrelin system and the development and progression of neuroendocrine tumors (NETs) is still controversial and remains unclear. METHODS Here, we systematically evaluated the expression levels (by quantitative-PCR) of key ghrelin system components of in gastroenteropancreatic (GEP)-NETs, as compared to non-tumor adjacent (NTA; n = 42) and normal tissues (NT; n = 14). Then, we analyzed their putative associations with clinical-histological characteristics. RESULTS The results indicate that ghrelin and its receptor GHSR1a are present in a high proportion of normal tissues, while the enzyme ghrelin-O-acyltransferase (GOAT) and the splicing variants In1-ghrelin and GHSR1b were present in a lower proportion of normal tissues. In contrast, all ghrelin system components were present in a high proportion of tumor and NTA tissues. GOAT was significantly overexpressed (by quantitative-PCR (qPCR)) in tumor samples compared to NTA, while a trend was found for ghrelin, In1-ghrelin and GHSR1a. In addition, expression of these components displayed significant correlations with key clinical parameters. The marked overexpression of GOAT in tumor samples compared to NTA regions was confirmed by IHC, revealing that this enzyme is particularly overexpressed in gastrointestinal NETs, where it is directly correlated with tumor diameter. CONCLUSIONS These results provide novel information on the presence and potential pathophysiological implications of the ghrelin system components in GEP-NETs, wherein GOAT might represent a novel diagnostic biomarker.
Collapse
Affiliation(s)
- Aura D Herrera-Martínez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Rafael Sánchez-Sánchez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Pathology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Emilia Alors-Perez
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Sergio Pedraza-Arevalo
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Raquel Serrano-Blanch
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Medical Oncology Service, Reina Sofia University Hospital, Córdoba, Spain
| | - Antonio J Martínez-Fuentes
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain.,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain.,Reina Sofia University Hospital, Córdoba, Spain
| | - Maria A Gálvez-Moreno
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Endocrinology and Nutrition Service, Reina Sofia University Hospital, Córdoba, Spain.
| | - Justo P Castaño
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute for Biomedical Research of Cordoba (IMIBIC), Córdoba, Spain. .,Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain. .,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Córdoba, Spain. .,Reina Sofia University Hospital, Córdoba, Spain.
| |
Collapse
|
16
|
Grönberg M, Nilsson C, Markholm I, Hedenfalk I, Blomqvist C, Holmberg L, Tiensuu Janson E, Fjällskog ML. Ghrelin expression is associated with a favorable outcome in male breast cancer. Sci Rep 2018; 8:13586. [PMID: 30206250 PMCID: PMC6134078 DOI: 10.1038/s41598-018-31783-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
Ghrelin and obestatin are two gastrointestinal peptides, derived from a common precursor. Expression of both peptides have been found in breast cancer tissue and ghrelin has been associated with breast cancer development. Ghrelin expression is associated with longer survival in women diagnosed with invasive and node negative breast cancer. The clinical implications of the peptide expression in male breast cancer are unclear. The aim of this study was to investigate the role and potential clinical value of ghrelin and obestatin in male breast cancer. A tissue microarray of invasive male breast cancer specimens from 197 patients was immunostained with antibodies versus the two peptides. The expression of the peptides was correlated to previously known prognostic factors in breast cancer and to the outcome. No strong correlations were found between ghrelin or obestatin expression and other known prognostic factors. Only ghrelin expression was statistically significantly correlated to breast cancer-specific survival (HR 0.39, 95% CI 0.18–0.83) in univariate analyses and in multivariate models, adjusted for tumor size and node status (HR 0.38, 95% CI 0.17–0.87). HR for obestatin was 0.38 (95% CI 0.11–1.24). Ghrelin is a potential prognostic factor for breast cancer death in male breast cancer. Patients with tumors expressing ghrelin have a 2.5-fold lower risk for breast cancer death than those lacking ghrelin expression. Drugs targeting ghrelin are currently being investigated in clinical studies treating metabolic or nutritional disorders. Ghrelin should be further evaluated in forthcoming studies as a prognostic marker with the aim to be included in decision algorithms.
Collapse
Affiliation(s)
- Malin Grönberg
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden.
| | - Cecilia Nilsson
- Center for Clinical Research, Västmanland County Hospital, Västerås, Sweden
| | - Ida Markholm
- Division of Oncology and Pathology, Department of Clinical Sciences, and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology and Pathology, Department of Clinical Sciences, and CREATE Health Strategic Center for Translational Cancer Research, Lund University, Lund, Sweden
| | - Carl Blomqvist
- Department of Oncology, Helsinki University, Helsinki, Finland.,Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Lars Holmberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.,Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Eva Tiensuu Janson
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | - Marie-Louise Fjällskog
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
17
|
Hou J, Kovacs MS, Dhanvantari S, Luyt LG. Development of Candidates for Positron Emission Tomography (PET) Imaging of Ghrelin Receptor in Disease: Design, Synthesis, and Evaluation of Fluorine-Bearing Quinazolinone Derivatives. J Med Chem 2018; 61:1261-1275. [DOI: 10.1021/acs.jmedchem.7b01754] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Jinqiang Hou
- London Regional Cancer Program, London N6A 4L6, Canada
- Lawson Health Research Institute, London N6C 2R5, Canada
| | | | | | - Leonard G. Luyt
- London Regional Cancer Program, London N6A 4L6, Canada
- Lawson Health Research Institute, London N6C 2R5, Canada
| |
Collapse
|
18
|
Charron CL, Hou J, McFarland MS, Dhanvantari S, Kovacs MS, Luyt LG. Structure–Activity Study of Ghrelin(1–8) Resulting in High Affinity Fluorine-Bearing Ligands for the Ghrelin Receptor. J Med Chem 2017; 60:7256-7266. [DOI: 10.1021/acs.jmedchem.7b00164] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Carlie L. Charron
- Department
of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
| | - Jinqiang Hou
- London
Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario N6A 4L6, Canada
| | - Mark S. McFarland
- Department
of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
| | - Savita Dhanvantari
- Imaging
Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
- Department
of Medical Biophysics, University of Western Ontario, 1151 Richmond
Street, London, Ontario N6A 5B7, Canada
| | - Michael S. Kovacs
- Imaging
Program, Lawson Health Research Institute, 268 Grosvenor Street, London, Ontario N6A 4V2, Canada
- Department
of Medical Biophysics, University of Western Ontario, 1151 Richmond
Street, London, Ontario N6A 5B7, Canada
| | - Leonard G. Luyt
- Department
of Chemistry, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A 5B7, Canada
- London
Regional Cancer Program, Lawson Health Research Institute, 790 Commissioners Road East, London, Ontario N6A 4L6, Canada
- Departments
of Oncology and Medical Imaging, University of Western Ontario, 1151
Richmond Street, London, Ontario N6A 5B7, Canada
| |
Collapse
|
19
|
Grönberg M, Ahlin C, Naeser Y, Janson ET, Holmberg L, Fjällskog ML. Ghrelin is a prognostic marker and a potential therapeutic target in breast cancer. PLoS One 2017; 12:e0176059. [PMID: 28419141 PMCID: PMC5395214 DOI: 10.1371/journal.pone.0176059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 04/04/2017] [Indexed: 12/26/2022] Open
Abstract
Ghrelin and obestatin are gastrointestinal peptides, encoded by the same preproghrelin gene. Both are expressed in breast cancer tissue and ghrelin has been implicated in breast cancer tumorigenesis. Despite recent advances in breast cancer management the need for new prognostic markers and potential therapeutic targets in breast cancer remains high. We studied the prognostic impact of ghrelin and obestatin in women with node negative breast cancer. Within a cohort of women with breast cancer with tumor size ≤ 50 mm, no lymph node metastases and no initiation of adjuvant chemotherapy, 190 women were identified who died from breast cancer and randomly selected 190 women alive at the corresponding time as controls. Tumor tissues were immunostained with antibodies versus the peptides. Ghrelin expression was associated with better breast cancer specific survival in univariate analyses (OR 0.55, 95% CI 0.36–0.84) and in multivariate models, adjusted for endocrine treatment and age (OR 0.57, 95% CI 0.36–0.89). Obestatin expression was non-informative (OR 1.2, 95% CI 0.60–2.46). Ghrelin expression is independent prognostic factor for breast cancer death in node negative patients—halving the risk for dying of breast cancer. Our data implies that ghrelin could be a potential therapeutic target in breast cancer treatment.
Collapse
Affiliation(s)
- Malin Grönberg
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
- * E-mail:
| | - Cecilia Ahlin
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Ylva Naeser
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | - Eva Tiensuu Janson
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| | - Lars Holmberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
- Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Marie-Louise Fjällskog
- Department of Medical Sciences, Section of Endocrine Oncology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Au CC, Furness JB, Brown KA. Ghrelin and Breast Cancer: Emerging Roles in Obesity, Estrogen Regulation, and Cancer. Front Oncol 2017; 6:265. [PMID: 28119851 PMCID: PMC5220482 DOI: 10.3389/fonc.2016.00265] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/12/2016] [Indexed: 01/26/2023] Open
Abstract
Local and systemic factors have been shown to drive the growth of breast cancer cells in postmenopausal obese women, who have increased risk of estrogen receptor-positive breast cancer. Estrogens, produced locally in the breast fat by the enzyme aromatase, have an important role in promoting cancer cell proliferation. Ghrelin, a 28-amino acid peptide hormone, may also influence cancer growth. This peptide is produced in the stomach and acts centrally to regulate appetite and growth hormone release. Circulating levels of ghrelin, and its unacylated form, des-acyl ghrelin, are almost always inversely correlated with obesity, and these peptide hormones have recently been shown to inhibit adipose tissue aromatase expression. Ghrelin and des-acyl ghrelin have also been shown to be produced by some tumor cells and influence tumor growth. The ghrelin/des-acyl ghrelin–cancer axis is complex, one reason being that tumor cells have been shown to express splice variants of ghrelin, and ghrelin and des-acyl ghrelin might act at receptors other than the cognate ghrelin receptor, growth hormone secretagogue receptor 1a, in tumors. Effects of ghrelin and des-acyl ghrelin on energy homeostasis may also affect tumor development and growth. This review will summarize our current understanding of the role of ghrelin and des-acyl ghrelin in hormone-dependent cancers, breast cancer in particular.
Collapse
Affiliation(s)
- CheukMan Cherie Au
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - John B Furness
- Department of Anatomy and Neuroscience, University of Melbourne and Florey Institute of Neuroscience and Mental Health , Parkville, VIC , Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia; Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
Prognostic Significance of Preoperative and Postoperative Plasma Levels of Ghrelin in Gastric Cancer: 3-Year Survival Study. Clin Transl Gastroenterol 2017; 8:e209. [PMID: 28055030 PMCID: PMC5288598 DOI: 10.1038/ctg.2016.64] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/21/2016] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVES: We aimed to investigate prognostic effects of plasma levels of ghrelin before and after gastrectomy in gastric cancer (GC). METHODS: We followed 81 GC patients up to 3 years in this study. They were candidates for curative gastrectomy with or without neoadjuvant chemotherapy. Plasma levels of total and active ghrelins before and after the operation were assessed. Association of plasma levels of ghrelin with survival were assessed and adjusted for other potential prognostic factors using Cox regression analyses. RESULTS: Both total and active ghrelins dropped after gastrectomy (P<0.001 for both). Multiple Cox models revealed worse survival for patients with postoperative total ghrelins below median (hazards ratio (HR)=2.33, 95% confidence interval (CI): 1.01–5.41) or 25th percentile (HR=4.29, 95% CI: 1.48–12.44) compared with patients with higher ghrelin levels. In case of preoperative total ghrelin, patients with either second or third quartiles of plasma ghrelin showed worse survival compared with patients with the lowest quartile (HR=2.67, 95% CI: 1.11–6.38 for second quartile, and HR=2.32, 95% CI: 1.01–5.35 for third quartile vs. the lowest quartile). However, there was no difference between patients with the highest and lowest quartiles (HR=0.78, 95% CI: 0.22–2.73). Similar pattern was observed for preoperative active ghrelin (HR=4.92, 95% CI: 1.80–13.54 for second quartile, and HR=2.87, 95% CI: 1.11–7.38 for third quartile vs. the lowest quartile). Advanced TNM stage (HR=4.88, 95% CI: 1.10–21.77), cachexia (HR=2.99, 95% CI: 1.35–6.63), and receiving no neoadjuvant chemotherapy (HR=2.02, 95% CI: 1.04–3.92) were other poor prognostic factors. CONCLUSIONS: Preoperative and postoperative plasma levels of ghrelin could predict survival of GC patients with different patterns. This prognostic effect was independent of stage and cachexia. Measurement of plasma ghrelin in GC patients could complement conventional staging for more precise risk-stratification of the patients. Extrinsic admirations of ghrelin after total gastrectomy has potentials to improve survival of GC patients.
Collapse
|
22
|
Li B, Zeng M, Zheng H, Huang C, He W, Lu G, Li X, Chen Y, Xie R. Effects of ghrelin on the apoptosis of human neutrophils in vitro. Int J Mol Med 2016; 38:794-802. [PMID: 27431014 PMCID: PMC4990324 DOI: 10.3892/ijmm.2016.2668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 06/30/2016] [Indexed: 01/19/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by lung inflammation and the diffuse infiltration of neutrophils into the alveolar space. Neutrophils are abundant, short-lived leukocytes that play a key role in immune defense against microbial infections. These cells die via apoptosis following the activation and uptake of microbes, and will also enter apoptosis spontaneously at the end of their lifespan if they do not encounter pathogens. Apoptosis is essential for the removal of neutrophils from inflamed tissues and for the timely resolution of neutrophilic inflammation. Ghrelin is an endogenous ligand for the growth hormone (GH) secretagogue receptor, produced and secreted mainly from the stomach. Previous studies have reported that ghrelin exerts anti-inflammatory effects in lung injury through the regulation of the apoptosis of different cell types; however, the ability of ghrelin to regulate alveolar neutrophil apoptosis remains largely undefined. We hypothesized that ghrelin may have the ability to modulate neutrophil apoptosis. In this study, to examine this hypothesis, we investigated the effects of ghrelin on freshly isolated neutrophils in vitro. Our findings demonstrated a decrease in the apoptotic ratio (as shown by flow cytometry), as well as in the percentage of cells with decreased mitochondrial membrane potential (ΔΨm) and in the terminal deoxynucleotidyl transferase (TdT)-mediated dUTP-biotin nick-end labeling-positive rate, accompanied by an increased B-cell lymphoma 2/Bax ratio and the downregulation of cleaved caspase-3 in neutrophils following exposure to lipopolysaccharide (100 ng/ml). However, pre-treatment with ghrelin at a physiological level (100 nM) did not have a notable influence on the neutrophils in all the aforementioned tests. Our findings suggest that ghrelin may not possess the ability to modulate the neutrophil lifespan in vitro.
Collapse
Affiliation(s)
- Bin Li
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Haichong Zheng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Chunrong Huang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Guifang Lu
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xia Li
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yanzhu Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Ruijie Xie
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
23
|
van Adrichem RCS, van der Lely AJ, Huisman M, Kramer P, Feelders RA, Delhanty PJD, de Herder WW. Plasma acylated and plasma unacylated ghrelin: useful new biomarkers in patients with neuroendocrine tumors? Endocr Connect 2016; 5:143-51. [PMID: 27215920 PMCID: PMC5002960 DOI: 10.1530/ec-16-0021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/23/2016] [Indexed: 12/18/2022]
Abstract
To date, the value of fasting plasma acylated ghrelin (AG) and unacylated ghrelin (UAG) as potential novel biomarkers in patients with neuroendocrine tumors (NETs) is unknown. The aims of this study are to (i) compare fasting AG and UAG levels between nonobese, nondiabetic NET patients (N=28) and age- (±3 years) and sex-matched nonobese, nondiabetic controls (N=28); and (ii) study the relationship between AG, UAG, and AG/UAG ratios and biochemical (chromogranin-A (CgA) and neuron-specific enolase (NSE) levels) and clinical parameters (age at diagnosis, sex, primary tumor location, carcinoid syndrome, ENETS TNM classification, Ki-67 proliferation index, grading, prior incomplete surgery) in NET patients. Fasting venous blood samples (N=56) were collected and directly stabilized with 4-(2-aminoethyl) benzenesulfonyl fluoride hydrochloride after withdrawal. Plasma AG and UAG levels were determined by ELISA. Expression of ghrelin was examined in tumor tissue by immunohistochemistry. There were no significant differences between NET patients and controls in AG (median: 62.5 pg/mL, IQR: 33.1-112.8 vs median: 57.2pg/mL, IQR: 26.7-128.3, P=0.66) and UAG in levels (median: 76.6pg/mL, IQR: 35.23-121.7 vs median: 64.9, IQR: 27.5-93.1, P=0.44). No significant correlations were found between AG, UAG, and AG/UAG ratios versus biochemical and clinical parameters in NET patients with the exception of age at diagnosis (AG: ρ= -0.47, P=0.012; AG/UAG ratio: ρ= -0.50, P=0.007) and baseline chromogranin-A levels (AG/UAG ratio: ρ= -0.44, P=0.019). In our view, fasting plasma acylated and unacylated ghrelin appear to have no value as diagnostic biomarkers in the clinical follow-up of patients with NETs.
Collapse
Affiliation(s)
- Roxanne C S van Adrichem
- Department of Internal MedicineSector of Endocrinology, ENETS Centre of Excellence for Neuroendocrine Tumors, Erasmus MC, Rotterdam, The Netherlands
| | | | - Martin Huisman
- Department of Internal MedicineErasmus MC, Rotterdam, The Netherlands
| | - Piet Kramer
- Department of Internal MedicineErasmus MC, Rotterdam, The Netherlands
| | - Richard A Feelders
- Department of Internal MedicineSector of Endocrinology, ENETS Centre of Excellence for Neuroendocrine Tumors, Erasmus MC, Rotterdam, The Netherlands
| | | | - Wouter W de Herder
- Department of Internal MedicineSector of Endocrinology, ENETS Centre of Excellence for Neuroendocrine Tumors, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Cecarini V, Bonfili L, Cuccioloni M, Keller JN, Bruce-Keller AJ, Eleuteri AM. Effects of Ghrelin on the Proteolytic Pathways of Alzheimer's Disease Neuronal Cells. Mol Neurobiol 2015; 53:3168-3178. [PMID: 26033219 DOI: 10.1007/s12035-015-9227-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/21/2015] [Indexed: 12/14/2022]
Abstract
Ghrelin is an orexigenic hormone with a role in the onset and progression of neurodegenerative disorders. It has been recently associated to Alzheimer's disease (AD) for its neuroprotective and anti-apoptotic activity. In the present study, we dissected the effect of ghrelin treatment on the two major intracellular proteolytic pathways, the ubiquitin-proteasome system (UPS) and autophagy, in cellular models of AD (namely SH-SY5Y neuroblastoma cells stably transfected with either the wild-type AβPP gene or the 717 valine-to-glycine AβPP-mutated gene). Ghrelin showed a growth-promoting effect on neuronal cells inducing also time-dependent modifications of the growth hormone secretagogue receptor type 1 (GHS-R1) expression. Interestingly, we demonstrated for the first time that ghrelin was able to activate the proteasome in neural cells playing also a role in the interplay between the UPS and autophagy. Our data provide a novel mechanism by which circulating hormones control neural homeostasis through the regulation of proteolytic pathways implicated in AD.
Collapse
Affiliation(s)
- Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy.
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| | - Jeffrey N Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Annadora J Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032, Camerino, Italy
| |
Collapse
|
25
|
Chen JA, Splenser A, Guillory B, Luo J, Mendiratta M, Belinova B, Halder T, Zhang G, Li YP, Garcia JM. Ghrelin prevents tumour- and cisplatin-induced muscle wasting: characterization of multiple mechanisms involved. J Cachexia Sarcopenia Muscle 2015; 6:132-43. [PMID: 26136189 PMCID: PMC4458079 DOI: 10.1002/jcsm.12023] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 11/11/2014] [Accepted: 02/13/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Cachexia and muscle atrophy are common consequences of cancer and chemotherapy administration. The novel hormone ghrelin has been proposed as a treatment for this condition. Increases in food intake and direct effects on muscle proteolysis and protein synthesis are likely to mediate these effects, but the pathways leading to these events are not well understood. METHODS We characterized molecular pathways involved in muscle atrophy induced by Lewis lung carcinoma (LLC) tumour implantation in c57/bl6 adult male mice and by administration of the chemotherapeutic agent cisplatin in mice and in C2C12 myotubes. The effects of exogenous ghrelin administration and its mechanisms of action were examined in these settings. RESULTS Tumour implantation and cisplatin induced muscle atrophy by activating pro-inflammatory cytokines, p38-C/EBP-β, and myostatin, and by down-regulating Akt, myoD, and myogenin, leading to activation of ubiquitin-proteasome-mediated proteolysis and muscle weakness. Tumour implantation also increased mortality. In vitro, cisplatin up-regulated myostatin and atrogin-1 by activating C/EBP-β and FoxO1/3. Ghrelin prevented these changes in vivo and in vitro, significantly increasing muscle mass (P < 0.05 for LLC and P < 0.01 for cisplatin models) and grip strength (P = 0.038 for LLC and P = 0.001 for cisplatin models) and improving survival (P = 0.021 for LLC model). CONCLUSION Ghrelin prevents muscle atrophy by down-regulating inflammation, p38/C/EBP-β/myostatin, and activating Akt, myogenin, and myoD. These changes appear, at least in part, to target muscle cells directly. Ghrelin administration in this setting is associated with improved muscle strength and survival.
Collapse
Affiliation(s)
- Ji-An Chen
- Department of Health Education, College of Preventive Medicine, Third Military Medical University, Chongqing, China.,Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Andres Splenser
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Bobby Guillory
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jiaohua Luo
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Department of Environmental Hygiene, College of Preventive Medicine, Third Military Medical University, Chongqing, China
| | - Meenal Mendiratta
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Blaga Belinova
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Tripti Halder
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Guohua Zhang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX, USA
| | - Jose M Garcia
- Division of Endocrinology, Diabetes and Metabolism, MCL, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Department of Medicine, Baylor College of Medicine, Houston, TX, USA.,Huffington Center on Aging and Dept. of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
26
|
Ghrelin improves functional survival of engrafted adipose-derived mesenchymal stem cells in ischemic heart through PI3K/Akt signaling pathway. BIOMED RESEARCH INTERNATIONAL 2015; 2015:858349. [PMID: 25879037 PMCID: PMC4387976 DOI: 10.1155/2015/858349] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) have been proposed as a promising cell population for cell therapy and regenerative medicine applications. However, the low retention and poor survival of engrafted cells hampered the therapeutic efficacy of engrafted MSCs. Ghrelin is a 28-amino-acid peptide hormone and is proved to exert a protective effect on the cardiovascular system. This study is designed to investigate the protective effects of ghrelin on engrafted adipose-derived mesenchymal stem cells (ADMSCs) and its beneficial effects with cellular therapy in mice myocardial infarction (MI). Results showed that intramyocardial injection of ADMSCs combining with ghrelin administration inhibited host cardiomyocyte apoptosis, reduced fibrosis, and improved cardiac function. To reveal possible mechanisms, ADMSCs were subjected to hypoxia/serum deprivation (H/SD) injury to simulate ischemic conditions in vivo. Ghrelin (10−8 M, 33712 pg/ml) improved ADMSCs survival under H/SD condition. Western blot assay revealed that ghrelin increased
AKT phosphorylation both in vivo and in vitro, decreased the proapoptotic protein Bax, and increased the antiapoptotic protein Bcl-2 in vitro, while these effects were abolished by PI3K inhibitor LY294002. These revealed that ghrelin may serve as a promising candidate for hormone-driven approaches to improve the efficacy of mesenchymal stem cell-based therapy for cardiac ischemic disease via PI3K/AKT pathway.
Collapse
|
27
|
Wang J, Guo S, Han L, Fang M, Wang L, Bartsch JW, Li J. Correlation of ghrelin and growth hormone secretagogue receptor expression with clinical features in human pituitary adenomas. Exp Ther Med 2015; 9:1909-1914. [PMID: 26136913 DOI: 10.3892/etm.2015.2341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 03/03/2015] [Indexed: 12/12/2022] Open
Abstract
Ghrelin, as a brain-gut peptide, has growth hormone (GH)-releasing and appetite-inducing activities and a widespread tissue distribution. Furthermore, ghrelin is an endogenous ligand of the GH secretagogue receptor (GHSR), and both ghrelin and GHSR are expressed in the pituitary; however, the data regarding the expression of ghrelin and GHSR in pituitary adenomas are divergent and conflicting. In the present study, therefore, the expression of ghrelin and GHSR was examined in the full spectrum of human pituitary adenoma subtypes (n=34) and in normal pituitary tissue (n=3). The mRNA and protein expression levels were quantified using a competitive reverse transcription-polymerase chain reaction and western blotting and the correlation of the results with the clinical parameters was assessed. mRNA and protein expression of ghrelin and GHSR was detected in all samples with the highest mean level in GH adenomas, a moderate level in clinically non-functioning adenomas and the lowest level in adrenocorticotropin adenomas. A significant correlation between the ghrelin and GHSR mRNA expression levels was observed in the GH adenomas (n=12) (r=0.8435, P=0.0006). The ghrelin mRNA expression level in the GH adenomas correlated positively with the basic serum GH level (n=12) (r=0.6488, P=0.0225). Furthermore, the mean level of ghrelin mRNA expression was significantly higher in invasive adenomas than in noninvasive adenomas (P<0.01). Collectively, the results of the study provided evidence that ghrelin and GHSR are expressed in the various subtypes of pituitary adenoma, with specific overexpression in GH adenomas. The study suggests that the binding of ghrelin to GHSR promotes the secretion of GH and plays an important role in the development of GH adenomas via autocrine and/or paracrine effects.
Collapse
Affiliation(s)
- Junwen Wang
- Department of Neurosurgery, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| | - Songbo Guo
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College Affiliated to Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Lin Han
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College Affiliated to Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Mingbo Fang
- Department of Neurosurgery, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| | - Lei Wang
- Department of Neurosurgery, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| | - Jörg W Bartsch
- Department of Neurosurgery, Marburg University, D-35033 Marburg, Germany
| | - Jun Li
- Department of Neurosurgery, Wuhan Central Hospital, Wuhan, Hubei 430014, P.R. China
| |
Collapse
|
28
|
The cardiovascular action of hexarelin. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2014; 11:253-8. [PMID: 25278975 PMCID: PMC4178518 DOI: 10.11909/j.issn.1671-5411.2014.03.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 05/25/2014] [Accepted: 07/10/2014] [Indexed: 11/21/2022]
Abstract
Hexarelin, a synthetic growth hormone-releasing peptide, can bind to and activate the growth hormone secretagogue receptor (GHSR) in the brain similar to its natural analog ghrelin. However, the peripheral distribution of GHSR in the heart and blood vessels suggests that hexarelin might have direct cardiovascular actions beyond growth hormone release and neuroendocrine effects. Furthermore, the non-GHSR CD36 had been demonstrated to be a specific cardiac receptor for hexarelin and to mediate its cardioprotective effects. When compared with ghrelin, hexarelin is chemically more stable and functionally more potent. Therefore, it may be a promising therapeutic agent for some cardiovascular conditions. In this concise review, we discuss the current evidence for the cardiovascular action of hexarelin.
Collapse
|
29
|
Wang L, Song Y, Li F, Liu Y, Ma J, Mao M, Wu F, Wu Y, Li S, Guan B, Liu X. Effects of Wen Dan Tang on insomnia-related anxiety and levels of the brain-gut peptide Ghrelin. Neural Regen Res 2014; 9:205-12. [PMID: 25206802 PMCID: PMC4146165 DOI: 10.4103/1673-5374.125351] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2013] [Indexed: 11/04/2022] Open
Abstract
Ghrelin, a brain-gut peptide that induces anxiety and other abnormal emotions, contributes to the effects of insomnia on emotional behavior. In contrast, the traditional Chinese Medicine remedy Wen Dan Tang reduces insomnia-related anxiety, which may perhaps correspond to changes in the brain-gut axis. This suggests a possible relationship between Wen Dan Tang's pharmacological mechanism and the brain-gut axis. Based on this hypothesis, a sleep-deprived rat model was induced and Wen Dan Tang was administered using oral gavage during model establishment. Wen Dan Tang significantly reduced insomnia-related anxiety and prevented Ghrelin level decreases following sleep deprivation, especially in the hypothalamus. Increased expression of Ghrelin receptor mRNA in the hypothalamus was also observed, suggesting that reduced anxiety may be a result of Wen Dan Tang's regulation of Ghrelin-Ghrelin receptors.
Collapse
Affiliation(s)
- Liye Wang
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Yuehan Song
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Feng Li
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Liu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Jie Ma
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Meng Mao
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Fengzhi Wu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Wu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Sinai Li
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Binghe Guan
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaolan Liu
- Nautical Chinese Medicine Key Subject, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Clinical development of ghrelin axis-derived molecules for cancer cachexia treatment. Curr Opin Support Palliat Care 2014; 7:368-75. [PMID: 24145681 DOI: 10.1097/spc.0000000000000012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Cachexia is a devastating complication of cancer for which there is no approved treatment. Here we review the clinical development of ghrelin and ghrelin mimetics (also known as growth hormone secretagogues or GHS) for cancer cachexia treatment. RECENT FINDINGS Ghrelin, a novel hormone known to increase appetite, lean and fat mass, and growth hormone secretion, is being developed as a therapeutic option for cancer anorexia-cachexia syndrome (CACS). Recent animal studies suggest that it may also decrease inflammation and that some of its effects may be independent of its only known receptor, the GHS receptor-1a.Clinical studies recently have shown that administration of ghrelin or GHS improves appetite and quality of life as assessed by questionnaires. Weight gain, increased food intake and better tolerance to chemotherapy have also been reported. This treatment appears to be safe and well tolerated. SUMMARY Ghrelin and GHS have the potential to effectively prevent or reverse CACS. Preliminary studies show improvements in weight stabilization and appetite with short-term usage. Further studies are required to fully characterize the role of ghrelin and GHS for the treatment of CACS and to establish the safety of this approach.
Collapse
|
31
|
Seim I, Jeffery PL, de Amorim L, Walpole CM, Fung J, Whiteside EJ, Lourie R, Herington AC, Chopin LK. Ghrelin O-acyltransferase (GOAT) is expressed in prostate cancer tissues and cell lines and expression is differentially regulated in vitro by ghrelin. Reprod Biol Endocrinol 2013; 11:70. [PMID: 23879975 PMCID: PMC3724588 DOI: 10.1186/1477-7827-11-70] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 07/05/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Ghrelin is a 28 amino acid peptide hormone that is expressed in the stomach and a range of peripheral tissues, where it frequently acts as an autocrine/paracrine growth factor. Ghrelin is modified by a unique acylation required for it to activate its cognate receptor, the growth hormone secretagogue receptor (GHSR), which mediates many of the actions of ghrelin. Recently, the enzyme responsible for adding the fatty acid residue (octanoyl/acyl group) to the third amino acid of ghrelin, GOAT (ghrelin O-acyltransferase), was identified. METHODS We used cell culture, quantitative real-time reverse transcription (RT)-PCR and immunohistochemistry to demonstrate the expression of GOAT in prostate cancer cell lines and tissues from patients. Real-time RT-PCR was used to demonstrate the expression of prohormone convertase (PC)1/3, PC2 and furin in prostate cancer cell lines. Prostate-derived cell lines were treated with ghrelin and desacyl ghrelin and the effect on GOAT expression was measured using quantitative RT-PCR. RESULTS We have demonstrated that GOAT mRNA and protein are expressed in the normal prostate and human prostate cancer tissue samples. The RWPE-1 and RWPE-2 normal prostate-derived cell lines and the LNCaP, DU145, and PC3 prostate cancer cell lines express GOAT and at least one other enzyme that is necessary to produce mature, acylated ghrelin from proghrelin (PC1/3, PC2 or furin). Finally, ghrelin, but not desacyl ghrelin (unacylated ghrelin), can directly regulate the expression of GOAT in the RWPE-1 normal prostate derived cell line and the PC3 prostate cancer cell line. Ghrelin treatment (100nM) for 6 hours significantly decreased GOAT mRNA expression two-fold (P < 0.05) in the PC3 prostate cancer cell line, however, ghrelin did not regulate GOAT expression in the DU145 and LNCaP prostate cancer cell lines. CONCLUSIONS This study demonstrates that GOAT is expressed in prostate cancer specimens and cell lines. Ghrelin regulates GOAT expression, however, this is likely to be cell-type specific. The expression of GOAT in prostate cancer supports the hypothesis that the ghrelin axis has autocrine/paracrine roles. We propose that the RWPE-1 prostate cell line and the PC3 prostate cancer cell line may be useful for investigating GOAT regulation and function.
Collapse
Affiliation(s)
- Inge Seim
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| | - Penny L Jeffery
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
- Mater Medical Research Institute, Mater Health Services, University of Queensland, South Brisbane, Queensland,, 4103, Australia
| | - Laura de Amorim
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Carina M Walpole
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Jenny Fung
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Eliza J Whiteside
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
| | - Rohan Lourie
- Mater Medical Research Institute, Mater Health Services, University of Queensland, South Brisbane, Queensland,, 4103, Australia
- Department of Pathology, Mater Health Services, South Brisbane, Queensland, 4103, Australia
| | - Adrian C Herington
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| | - Lisa K Chopin
- Ghrelin Research Group, Translational Research Institute - Institute of Health and Biomedical Innovation, Queensland University of Technology, 37 Kent St, Woolloongabba, Queensland, 4102, Australia
- Australian Prostate Cancer Research Centre, Queensland, Princess Alexandra Hospital, 199 Ipswich Road, Brisbane, Queensland, 4102, Australia
| |
Collapse
|
32
|
Abstract
Ingestion of food affects the secretion of hormones from specialized endocrine cells scattered within the intestinal mucosa. Upon release, these hormones mostly decrease food intake by signaling information to the brain. Although enteroendocrine cells in the small intestine were thought to represent the predominant gut-brain regulators of food intake, recent advances also established a major role for gastric hormones in these regulatory pathways. First and foremost, the gastric endocrine X/A-like cell was in the focus of many studies due to the production of ghrelin, which is until now the only known orexigenic hormone that is peripherally produced and centrally acting. Although X/A-cells were initially thought to only release one hormone that stimulates food intake, this view has changed with the identification of additional peptide products also derived from this cell, namely desacyl ghrelin, obestatin, and nesfatin-1. Desacyl ghrelin may play a counter-regulatory role to the food intake stimulatory effect of ghrelin. The same property was suggested for obestatin; however, this hypothesis could not be confirmed in numerous subsequent studies. Moreover, the description of the stomach as the major source of the novel anorexigenic hormone nesfatin-1 derived from the NUCB2 gene further corroborated the assumption that the gastric X/A-like cell products are not only stimulant but also inhibitors of feeding, thereby acting as so far unique dual regulator of food intake located in a logistically important place where the gastrointestinal tract has initial contact with food.
Collapse
Affiliation(s)
- Andreas Stengel
- Charité Center for Internal Medicine and Dermatology, Division Psychosomatic Medicine, Obesity Center Berlin, Charité Universitätsmedizin Berlin, Campus Mitte, Charitéplatz 1, 10117 Berlin, Germany.
| | | |
Collapse
|
33
|
Smitka K, Papezova H, Vondra K, Hill M, Hainer V, Nedvidkova J. The role of "mixed" orexigenic and anorexigenic signals and autoantibodies reacting with appetite-regulating neuropeptides and peptides of the adipose tissue-gut-brain axis: relevance to food intake and nutritional status in patients with anorexia nervosa and bulimia nervosa. Int J Endocrinol 2013; 2013:483145. [PMID: 24106499 PMCID: PMC3782835 DOI: 10.1155/2013/483145] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/11/2013] [Indexed: 12/13/2022] Open
Abstract
Eating disorders such as anorexia (AN) and bulimia nervosa (BN) are characterized by abnormal eating behavior. The essential aspect of AN is that the individual refuses to maintain a minimal normal body weight. The main features of BN are binge eating and inappropriate compensatory methods to prevent weight gain. The gut-brain-adipose tissue (AT) peptides and neutralizing autoantibodies play an important role in the regulation of eating behavior and growth hormone release. The mechanisms for controlling food intake involve an interplay between gut, brain, and AT. Parasympathetic, sympathetic, and serotoninergic systems are required for communication between brain satiety centre, gut, and AT. These neuronal circuits include neuropeptides ghrelin, neuropeptide Y (NPY), peptide YY (PYY), cholecystokinin (CCK), leptin, putative anorexigen obestatin, monoamines dopamine, norepinephrine (NE), serotonin, and neutralizing autoantibodies. This extensive and detailed report reviews data that demonstrate that hunger-satiety signals play an important role in the pathogenesis of eating disorders. Neuroendocrine dysregulations of the AT-gut-brain axis peptides and neutralizing autoantibodies may result in AN and BN. The circulating autoantibodies can be purified and used as pharmacological tools in AN and BN. Further research is required to investigate the orexigenic/anorexigenic synthetic analogs and monoclonal antibodies for potential treatment of eating disorders in clinical practice.
Collapse
Affiliation(s)
- Kvido Smitka
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Hana Papezova
- Psychiatric Clinic, First Faculty of Medicine, Charles University, Ke Karlovu 11, 121 08 Prague 2, Czech Republic
| | - Karel Vondra
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Martin Hill
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Vojtech Hainer
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
| | - Jara Nedvidkova
- Institute of Endocrinology, Laboratory of Clinical and Experimental Neuroendocrinology, Narodni 8, 116 94 Prague 1, Czech Republic
- *Jara Nedvidkova:
| |
Collapse
|
34
|
Lee SY, Sohn KA, Kim JH. MicroRNA-centric measurement improves functional enrichment analysis of co-expressed and differentially expressed microRNA clusters. BMC Genomics 2012; 13 Suppl 7:S17. [PMID: 23281707 PMCID: PMC3521213 DOI: 10.1186/1471-2164-13-s7-s17] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background Functional annotations are available only for a very small fraction of microRNAs (miRNAs) and very few miRNA target genes are experimentally validated. Therefore, functional analysis of miRNA clusters has typically relied on computational target gene prediction followed by Gene Ontology and/or pathway analysis. These previous methods share the limitation that they do not consider the many-to-many-to-many tri-partite network topology between miRNAs, target genes, and functional annotations. Moreover, the highly false-positive nature of sequence-based target prediction algorithms causes propagation of annotation errors throughout the tri-partite network. Results A new conceptual framework is proposed for functional analysis of miRNA clusters, which extends the conventional target gene-centric approaches to a more generalized tri-partite space. Under this framework, we construct miRNA-, target link-, and target gene-centric computational measures incorporating the whole tri-partite network topology. Each of these methods and all their possible combinations are evaluated on publicly available miRNA clusters and with a wide range of variations for miRNA-target gene relations. We find that the miRNA-centric measures outperform others in terms of the average specificity and functional homogeneity of the GO terms significantly enriched for each miRNA cluster. Conclusions We propose novel miRNA-centric functional enrichment measures in a conceptual framework that connects the spaces of miRNAs, genes, and GO terms in a unified way. Our comprehensive evaluation result demonstrates that functional enrichment analysis of co-expressed and differentially expressed miRNA clusters can substantially benefit from the proposed miRNA-centric approaches.
Collapse
Affiliation(s)
- Su Yeon Lee
- Seoul National University Biomedical Informatics (SNUBI) and Systems Biomedical Informatics Research Center, Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul 110799, Korea
| | | | | |
Collapse
|
35
|
Majchrzak K, Pawłowski KM, Orzechowska EJ, Dolka I, Mucha J, Motyl T, Król M. A role of ghrelin in canine mammary carcinoma cells proliferation, apoptosis and migration. BMC Vet Res 2012; 8:170. [PMID: 22999388 PMCID: PMC3514346 DOI: 10.1186/1746-6148-8-170] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 09/20/2012] [Indexed: 11/13/2022] Open
Abstract
Background Ghrelin is a natural ligand of the growth hormone secretagogue receptor (GHS-R). They are often co-expressed in multiple human tumors and related cancer cell lines what can indicate that the ghrelin/GHS-R axis may have an important role in tumor growth and progression. However, a role of ghrelin in canine tumors remains unknown. Thus, the aim of our study was two-fold: (1) to assess expression of ghrelin and its receptor in canine mammary cancer and (2) to examine the effect of ghrelin on carcinoma cells proliferation, apoptosis, migration and invasion. The expression of ghrelin and its receptor in canine mammary cancer tissues and cell lines (isolated from primary tumors and their metastases) was examined using Real-time qPCR and immunohistochemistry. For apoptosis analysis the Annexin V and propidium iodide dual staining was applied whereas cell proliferation was evaluated by MTT assay and BrdU incorporation test. The influence of ghrelin on cancer cells migration and invasion was assessed using Boyden chamber assays and wound healing assay. Results The highest expression of ghrelin was observed in metastatic cancers whereas the lowest expression of ghrelin receptor was detected in tumors of the 3rd grade of malignancy. Higher expression of ghrelin and its receptor was detected in cancer cell lines isolated from metastases than in cell lines isolated from primary tumors. In vitro experiments demonstrated that exposure to low doses of ghrelin stimulates cellular proliferation, inhibits apoptosis and promotes motility and invasion of canine mammary cancer cells. Growth hormone secretagogue receptor inhibitor ([D-Lys3]-GHRP6) as well as RNA interference enhances early apoptosis. Conclusion The presence of ghrelin and GHS-R in all of the examined canine mammary tumors may indicate their biological role in cancer growth and development. Our experiments conducted in vitro confirmed that ghrelin promotes cancer development and metastasis.
Collapse
Affiliation(s)
- Kinga Majchrzak
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences - WULS, Nowoursynowska 159, Warsaw, 02-776, Poland
| | | | | | | | | | | | | |
Collapse
|
36
|
Kontoravdis N, Vassilikostas G, Lagoudianakis E, Pappas A, Seretis C, Panagiotopoulos N, Koronakis N, Chrysikos J, Karanikas G, Manouras I, Legakis I, Voros D. Effect of Acute Surgical Stress on Serum Ghrelin Levels. Gastroenterology Res 2012; 5:97-102. [PMID: 27785188 PMCID: PMC5051122 DOI: 10.4021/gr455e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2012] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Ghrelin is an appetite hormone that influences the gastrointestinal function and regulates energy metabolism. Growing evidence also suggests that this hormone plays a central role in immune modulation. Each surgical operation is followed by a series of inflammatory and metabolic changes that constitute the stress response. The aim of our study is to evaluate the effect of stress during different types of abdominal surgery in ghrelin serum levels. METHODS An overall of 25 patients were prospectively allocated in two groups based on the type of surgical operation. Group A (n = 10) patients were scheduled to undergo cholecystectomy, whereas Group B (n = 15) patients underwent colectomy. Serum ghrelin concentrations were evaluated in each patient preoperatively, after the induction of general anesthesia and tracheal intubation, one and five hours after the beginning of surgery and the morning of the first and second postoperative day. RESULTS In both groups serum ghrelin concentrations reached their peak level at 24 hr (Group A: 8.4 ± 3.4 ng/mL; Group B: 7.4 ± 1.8 ng/mL) and these values were significantly higher than those in the preoperative period (Group A: 5.0 ±1.5 ng/mL; Group B: 4.8 ± 0.6 ng/mL) (P < 0.05). Forty eight hours after surgery the levels of ghrelin returned to their preoperative status. Patients' gender, age, ASA score and type of surgical procedure did not influence the serum ghrelin levels. CONCLUSIONS Serum ghrelin concentration appears to elevate in response to surgical stress. Future studies are needed to improve comprehension of the mechanisms underlying responses of this hormone to acute surgical stress and to evaluate their possible clinical implications.
Collapse
Affiliation(s)
- Nikolaos Kontoravdis
- Second Department of Surgery, Aretaieion University Hospital, University of Athens, Greece
| | - George Vassilikostas
- Second Department of Surgery, Aretaieion University Hospital, University of Athens, Greece
| | | | - Apostolos Pappas
- 1st Department of Propaedeutic Surgery, Hippokrateion Hospital, Athens Medical School, University of Athens, Q. Sofias 114 avenue, 11527 Athens, Greece
| | | | | | - Nikolaos Koronakis
- Second Department of Surgery, 417 NIMTS (Military Veterans' Fund Hospital), Athens, Greece
| | - John Chrysikos
- Second Department of Surgery, 417 NIMTS (Military Veterans' Fund Hospital), Athens, Greece
| | - George Karanikas
- Second Department of Surgery, 417 NIMTS (Military Veterans' Fund Hospital), Athens, Greece
| | - Ioannis Manouras
- 1st Department of Propaedeutic Surgery, Hippokrateion Hospital, Athens Medical School, University of Athens, Q. Sofias 114 avenue, 11527 Athens, Greece
| | - Ioanis Legakis
- Department of Endocrinology, Henry Dunant Hospital, Athens, Greece
| | - Dionysios Voros
- Second Department of Surgery, Aretaieion University Hospital, University of Athens, Greece
| |
Collapse
|
37
|
Kim YS, Lee JS, Lee TH, Cho JY, Kim JO, Kim WJ, Kim HG, Jeon SR, Jeong HS. Plasma levels of acylated ghrelin in patients with functional dyspepsia. World J Gastroenterol 2012; 18:2231-7. [PMID: 22611317 PMCID: PMC3351774 DOI: 10.3748/wjg.v18.i18.2231] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 09/20/2011] [Accepted: 03/10/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the relationship between plasma acylated ghrelin levels and the pathophysiology of functional dyspepsia.
METHODS: Twenty-two female patients with functional dyspepsia and twelve healthy volunteers were recruited for the study. The functional dyspepsia patients were each diagnosed based on the Rome III criteria. Eligible patients completed a questionnaire concerning the severity of 10 symptoms. Plasma acylated ghrelin levels before and after a meal were determined in the study participants using a commercial human acylated enzyme immunoassay kit; electrogastrograms were performed for 50 min before and after a standardized 10-min meal containing 265 kcal.
RESULTS: There were no significant differences in plasma acylated ghrelin levels between healthy volunteers and patients with functional dyspepsia. However, in patients with functional dyspepsia, there was a negative correlation between fasting plasma acylated ghrelin levels and the sum score of epigastric pain (r = -0.427, P = 0.047) and a positive correlation between the postprandial/fasting plasma acylated ghrelin ratio and the sum score of early satiety (r = 0.428, P =0.047). Additionally, there was a negative correlation between fasting acylated ghrelin plasma levels and fasting normogastria (%) (r = -0.522, P = 0.013). Interestingly, two functional dyspepsia patients showed paradoxically elevated plasma acylated ghrelin levels after the meal.
CONCLUSION: Abnormal plasma acylated ghrelin levels before or after a meal may be related to several of the dyspeptic symptoms seen in patients with functional dyspepsia.
Collapse
|
38
|
Stengel A, Taché Y. Yin and Yang - the Gastric X/A-like Cell as Possible Dual Regulator of Food Intake. J Neurogastroenterol Motil 2012; 18:138-49. [PMID: 22523723 PMCID: PMC3325299 DOI: 10.5056/jnm.2012.18.2.138] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Accepted: 03/11/2012] [Indexed: 01/14/2023] Open
Abstract
Ingestion of food affects secretion of hormones from enteroendocrine cells located in the gastrointestinal mucosa. These hormones are involved in the regulation of various gastrointestinal functions including the control of food intake. One cell in the stomach, the X/A-like has received much attention over the past years due to the production of ghrelin. Until now, ghrelin is the only known orexigenic hormone that is peripherally produced and centrally acting to stimulate food intake. Subsequently, additional peptide products of this cell have been described including desacyl ghrelin, obestatin and nesfatin-1. Desacyl ghrelin seems to be involved in the regulation of food intake as well and could play a counter-balancing role of ghrelin's orexigenic effect. In contrast, the initially proposed anorexigenic action of obestatin did not hold true and therefore the involvement of this peptide in the regulation of feeding is questionable. Lastly, the identification of nesfatin-1 in the same cell in different vesicles than ghrelin extended the function of this cell type to the inhibition of feeding. Therefore, this X/A-like cell could play a unique role by encompassing yin and yang properties to mediate not only hunger but also satiety.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, Division Psychosomatic Medicine and Psychotherapy, Charité, Campus Mitte, Universitätsmedizin Berlin, Berlin, Germany
| | | |
Collapse
|
39
|
Growth hormone secretagogues protect mouse cardiomyocytes from in vitro ischemia/reperfusion injury through regulation of intracellular calcium. PLoS One 2012; 7:e35265. [PMID: 22493744 PMCID: PMC3320867 DOI: 10.1371/journal.pone.0035265] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 03/14/2012] [Indexed: 01/08/2023] Open
Abstract
Background Ischemic heart disease is a leading cause of mortality. To study this disease, ischemia/reperfusion (I/R) models are widely used to mimic the process of transient blockage and subsequent recovery of cardiac coronary blood supply. We aimed to determine whether the presence of the growth hormone secretagogues, ghrelin and hexarelin, would protect/improve the function of heart from I/R injury and to examine the underlying mechanisms. Methodology/Principal Findings Isolated hearts from adult male mice underwent 20 min global ischemia and 30 min reperfusion using a Langendorff apparatus. Ghrelin (10 nM) or hexarelin (1 nM) was introduced into the perfusion system either 10 min before or after ischemia, termed pre- and post-treatments. In freshly isolated cardiomyocytes from these hearts, single cell shortening, intracellular calcium ([Ca2+]i) transients and caffeine-releasable sarcoplasmic reticulum (SR) Ca2+ were measured. In addition, RT-PCR and Western blots were used to examine the expression level of GHS receptor type 1a (GHS-R1a), and phosphorylated phospholamban (p-PLB), respectively. Ghrelin and hexarelin pre- or post-treatments prevented the significant reduction in the cell shortening, [Ca2+]i transient amplitude and caffeine-releasable SR Ca2+ content after I/R through recovery of p-PLB. GHS-R1a antagonists, [D-Lys3]-GHRP-6 (200 nM) and BIM28163 (100 nM), completely blocked the effects of GHS on both cell shortening and [Ca2+]i transients. Conclusion/Significance Through activation of GHS-R1a, ghrelin and hexarelin produced a positive inotropic effect on ischemic cardiomyocytes and protected them from I/R injury probably by protecting or recovering p-PLB (and therefore SR Ca2+ content) to allow the maintenance or recovery of normal cardiac contractility. These observations provide supporting evidence for the potential therapeutic application of ghrelin and hexarelin in patients with cardiac I/R injury.
Collapse
|
40
|
Fung JNT, Seim I, Wang D, Obermair A, Chopin LK, Chen C. Expression and in vitro functions of the ghrelin axis in endometrial cancer. Discov Oncol 2011; 1:245-55. [PMID: 21761369 DOI: 10.1007/s12672-010-0047-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Ghrelin is a peptide hormone produced in the stomach and a range of other tissues, where it has endocrine, paracrine and autocrine roles in both normal and disease states. Ghrelin has been shown to be an important growth factor for a number of tumours, including prostate and breast cancers. In this study, we examined the expression of the ghrelin axis (ghrelin and its receptor, the growth hormone secretagogue receptor, GHSR) in endometrial cancer. Ghrelin is expressed in a range of endometrial cancer tissues, while its cognate receptor, GHSR1a, is expressed in a small subset of normal and cancer tissues. Low to moderately invasive endometrial cancer cell lines were examined by RT-PCR and immunoblotting, demonstrating that ghrelin axis mRNA and protein expression correlate with differentiation status of Ishikawa, HEC1B and KLE endometrial cancer cell lines. Moreover, treatment with ghrelin potently stimulated cell proliferation and inhibited cell death. Taken together, these data indicate that ghrelin promotes the progression of endometrial cancer cells in vitro, and may contribute to endometrial cancer pathogenesis and represent a novel treatment target.
Collapse
Affiliation(s)
- Jenny N T Fung
- Department of Physiology and Pharmacology, School of Biomedical Sciences, The University of Queensland, Brisbane QLD 4072, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Frago LM, Baquedano E, Argente J, Chowen JA. Neuroprotective actions of ghrelin and growth hormone secretagogues. Front Mol Neurosci 2011; 4:23. [PMID: 21994488 PMCID: PMC3182030 DOI: 10.3389/fnmol.2011.00023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Accepted: 08/29/2011] [Indexed: 11/21/2022] Open
Abstract
The brain incorporates and coordinates information based on the hormonal environment, receiving information from peripheral tissues through the circulation. Although it was initially thought that hormones only acted on the hypothalamus to perform endocrine functions, it is now known that they in fact exert diverse actions on many different brain regions including the hypothalamus. Ghrelin is a gastric hormone that stimulates growth hormone secretion and food intake to regulate energy homeostasis and body weight by binding to its receptor, growth hormone secretagogues–GH secretagogue-receptor, which is most highly expressed in the pituitary and hypothalamus. In addition, ghrelin has effects on learning and memory, reward and motivation, anxiety, and depression, and could be a potential therapeutic agent in neurodegenerative disorders where excitotoxic neuronal cell death and inflammatory processes are involved.
Collapse
Affiliation(s)
- Laura M Frago
- Department of Pediatrics, Universidad Autónoma de Madrid Madrid, Spain
| | | | | | | |
Collapse
|
42
|
Gahete MD, Córdoba-Chacón J, Hergueta-Redondo M, Martínez-Fuentes AJ, Kineman RD, Moreno-Bueno G, Luque RM, Castaño JP. A novel human ghrelin variant (In1-ghrelin) and ghrelin-O-acyltransferase are overexpressed in breast cancer: potential pathophysiological relevance. PLoS One 2011; 6:e23302. [PMID: 21829727 PMCID: PMC3150424 DOI: 10.1371/journal.pone.0023302] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 07/14/2011] [Indexed: 12/02/2022] Open
Abstract
The human ghrelin gene, which encodes the ghrelin and obestatin peptides, contains 5 exons (Ex), with Ex1-Ex4 encoding a 117 amino-acid (aa) preproprotein that is known to be processed to yield a 28-aa (ghrelin) and/or a 23-aa (obestatin) mature peptides, which possess biological activities in multiple tissues. However, the ghrelin gene also encodes additional peptides through alternative splicing or post-translational modifications. Indeed, we previously identified a spliced mRNA ghrelin variant in mouse (In2-ghrelin-variant), which is regulated in a tissue-dependent manner by metabolic status and may thus be of biological relevance. Here, we have characterized a new human ghrelin variant that contains Ex0-1, intron (In) 1, and Ex2 and lacks Ex3-4. This human In1-ghrelin variant would encode a new prepropeptide that conserves the first 12aa of native-ghrelin (including the Ser3-potential octanoylation site) but has a different C-terminal tail. Expression of In1-variant was detected in 22 human tissues and its levels were positively correlated with those of ghrelin-O-acyltransferase (GOAT; p = 0.0001) but not with native-ghrelin expression, suggesting that In1-ghrelin could be a primary substrate for GOAT in human tissues. Interestingly, levels of In1-ghrelin variant expression in breast cancer samples were 8-times higher than those of normal mammary tissue, and showed a strong correlation in breast tumors with GOAT (p = 0.0001), ghrelin receptor-type 1b (GHSR1b; p = 0.049) and cyclin-D3 (a cell-cycle inducer/proliferation marker; p = 0.009), but not with native-ghrelin or GHSR1a expression. Interestingly, In1-ghrelin variant overexpression increased basal proliferation of MDA-MB-231 breast cancer cells. Taken together, our results provide evidence that In1-ghrelin is a novel element of the ghrelin family with a potential pathophysiological role in breast cancer.
Collapse
Affiliation(s)
- Manuel D. Gahete
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Hospital Universitario Reina Sofía, and CIBERobn Fisiopatología de la Obesidad y la Nutrición, Córdoba, Spain
| | - José Córdoba-Chacón
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Hospital Universitario Reina Sofía, and CIBERobn Fisiopatología de la Obesidad y la Nutrición, Córdoba, Spain
| | - Marta Hergueta-Redondo
- Department of Biochemistry, Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPAZ) and Fundación MD Anderson Internacional, Madrid, Spain
| | - Antonio J. Martínez-Fuentes
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Hospital Universitario Reina Sofía, and CIBERobn Fisiopatología de la Obesidad y la Nutrición, Córdoba, Spain
| | - Rhonda D. Kineman
- Section of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, and Research and Development Division, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, United States of America
| | - Gema Moreno-Bueno
- Department of Biochemistry, Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC-UAM, Instituto de Investigación Sanitaria La Paz (IdiPAZ) and Fundación MD Anderson Internacional, Madrid, Spain
| | - Raúl M. Luque
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Hospital Universitario Reina Sofía, and CIBERobn Fisiopatología de la Obesidad y la Nutrición, Córdoba, Spain
- * E-mail: (JPC); (RML)
| | - Justo P. Castaño
- Department of Cell Biology, Physiology and Immunology, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), University of Córdoba, Hospital Universitario Reina Sofía, and CIBERobn Fisiopatología de la Obesidad y la Nutrición, Córdoba, Spain
- * E-mail: (JPC); (RML)
| |
Collapse
|
43
|
Stengel A, Taché Y. Interaction between gastric and upper small intestinal hormones in the regulation of hunger and satiety: ghrelin and cholecystokinin take the central stage. Curr Protein Pept Sci 2011; 12:293-304. [PMID: 21428875 PMCID: PMC3670092 DOI: 10.2174/138920311795906673] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 03/23/2011] [Indexed: 12/13/2022]
Abstract
Several peptides are produced and released from endocrine cells scattered within the gastric oxyntic and the small intestinal mucosa. These peptide hormones are crucially involved in the regulation of gastrointestinal functions and food intake by conveying their information to central regulatory sites located in the brainstem as well as in the forebrain, such as hypothalamic nuclei. So far, ghrelin is the only known hormone that is peripherally produced in gastric X/A-like cells and centrally acting to stimulate food intake, whereas the suppression of feeding seems to be much more redundantly controlled by a number of gut peptides. Cholecystokinin produced in the duodenum is a well established anorexigenic hormone that interacts with ghrelin to modulate food intake indicating a regulatory network located at the first site of contact with nutrients in the stomach and upper small intestine. In addition, a number of peptides including leptin, urocortin 2, amylin and glucagon-like peptide 1 interact synergistically with CCK to potentiate its satiety signaling effect. New developments have led to the identification of additional peptides in X/A-like cells either derived from the pro-ghrelin gene by alternative splicing and posttranslational processing (obestatin) or a distinct gene (nucleobindin2/nesfatin-1) which have been investigated for their influence on food intake.
Collapse
Affiliation(s)
- Andreas Stengel
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Yvette Taché
- Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division UCLA, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| |
Collapse
|
44
|
Arranz A, Venihaki M, Mol B, Androulidaki A, Dermitzaki E, Rassouli O, Ripoll J, Stathopoulos EN, Gomariz RP, Margioris AN, Tsatsanis C. The impact of stress on tumor growth: peripheral CRF mediates tumor-promoting effects of stress. Mol Cancer 2010; 9:261. [PMID: 20875132 PMCID: PMC2956730 DOI: 10.1186/1476-4598-9-261] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 09/27/2010] [Indexed: 11/20/2022] Open
Abstract
Introduction Stress has been shown to be a tumor promoting factor. Both clinical and laboratory studies have shown that chronic stress is associated with tumor growth in several types of cancer. Corticotropin Releasing Factor (CRF) is the major hypothalamic mediator of stress, but is also expressed in peripheral tissues. Earlier studies have shown that peripheral CRF affects breast cancer cell proliferation and motility. The aim of the present study was to assess the significance of peripheral CRF on tumor growth as a mediator of the response to stress in vivo. Methods For this purpose we used the 4T1 breast cancer cell line in cell culture and in vivo. Cells were treated with CRF in culture and gene specific arrays were performed to identify genes directly affected by CRF and involved in breast cancer cell growth. To assess the impact of peripheral CRF as a stress mediator in tumor growth, Balb/c mice were orthotopically injected with 4T1 cells in the mammary fat pad to induce breast tumors. Mice were subjected to repetitive immobilization stress as a model of chronic stress. To inhibit the action of CRF, the CRF antagonist antalarmin was injected intraperitoneally. Breast tissue samples were histologically analyzed and assessed for neoangiogenesis. Results Array analysis revealed among other genes that CRF induced the expression of SMAD2 and β-catenin, genes involved in breast cancer cell proliferation and cytoskeletal changes associated with metastasis. Cell transfection and luciferase assays confirmed the role of CRF in WNT- β-catenin signaling. CRF induced 4T1 cell proliferation and augmented the TGF-β action on proliferation confirming its impact on TGFβ/SMAD2 signaling. In addition, CRF promoted actin reorganization and cell migration, suggesting a direct tumor-promoting action. Chronic stress augmented tumor growth in 4T1 breast tumor bearing mice and peripheral administration of the CRF antagonist antalarmin suppressed this effect. Moreover, antalarmin suppressed neoangiogenesis in 4T1 tumors in vivo. Conclusion This is the first report demonstrating that peripheral CRF, at least in part, mediates the tumor-promoting effects of stress and implicates CRF in SMAD2 and β-catenin expression.
Collapse
Affiliation(s)
- Alicia Arranz
- Department of Clinical Chemistry, School Of Medicine, University of Crete, 71003 Heraklion, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu Z, Yan Y, Liu S, Wang F, Chen X. (18)F, (64)Cu, and (68)Ga labeled RGD-bombesin heterodimeric peptides for PET imaging of breast cancer. Bioconjug Chem 2010; 20:1016-25. [PMID: 20540537 DOI: 10.1021/bc9000245] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Radiolabeled RGD (Arg-Gly-Asp) and bombesin (BBN) radiotracers that specifically target integrin alpha(v)beta(3) and gastrin releasing peptide receptor (GRPR) are both promising radiopharmaceuticals for tumor imaging. We recently designed and synthesized a RGD-BBN heterodimeric peptide with both RGD and BBN motifs in one single molecule. The (18)F-labeled RGD-BBN heterodimer exhibited dual integrin alpha(v)beta(3) and GRPR targeting in a PC-3 prostate cancer model. In this study we investigated whether radiolabeled RGD-BBN tracers can be used to detect breast cancer by using microPET. Cell binding assay demonstrated that the high GRPR expressing breast cancer cells typically express low to moderate level of integrin alpha(v)beta(3), while high integrin alpha(v)beta(3) expressing breast cancer cells have negligible level of GRPR. We labeled RGD-BBN heterodimer with three positron emitting radionuclides (18)F, (64)Cu, and (68)Ga and investigated the corresponding PET radiotracers in both orthotopic T47D (GRPR(+)/low integrin alpha(v)beta(3)) and MDA-MB-435 (GRPR(-)/integrin alpha(v)beta(3)(+)) breast cancer models. The three radiotracers all possessed in vitro dual integrin alpha(v)beta(3) and GRPR binding affinity. The advantages of the RGD-BBN radiotracers over the corresponding BBN analogues are obvious for imaging MDA-MB-435 (GRPR(-)/integrin alpha(v)beta(3)(+)) tumor. (18)F-FB-PEG(3)-RGD-BBN showed lower tumor uptake than (64)Cu-NOTA-RGD-BBN and (68)Ga-NOTA-RGD-BBN but was able to visualize breast cancer tumors with high contrast. Synthesis of (64)Cu-NOTA-RGD-BBN and (68)Ga-NOTA-RGD-BBN is much faster and easier than (18)F-FB-PEG(3)-RGD-BBN. (64)Cu-NOTA-RGD-BBN showed prolonged tumor uptake but also higher liver retention and kidney uptake than (68)Ga-NOTA-RGD-BBN and (18)F-FB-PEG(3)-RGD-BBN. (68)Ga-NOTA-RGD-BBN possessed high tumor signals but also relatively high background uptake compared with the other two radiotracers. In summary, the prosthetic labeling groups, chelators, and isotopes all have a profound effect on the tumor targeting efficacy and in vivo kinetics of the RGD-BBN tracers for dual integrin and GRPR recognition. Further development of suitably labeled RGD-BBN tracers for PET imaging of cancer is warranted.
Collapse
Affiliation(s)
- Zhaofei Liu
- Department of Radiology, Biophysics, Stanford University School of Medicine, Stanford, CA94305-5484, USA
| | | | | | | | | |
Collapse
|
46
|
Ferrini F, Salio C, Lossi L, Merighi A. Ghrelin in central neurons. Curr Neuropharmacol 2010; 7:37-49. [PMID: 19721816 PMCID: PMC2724662 DOI: 10.2174/157015909787602779] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 08/15/2008] [Accepted: 09/01/2008] [Indexed: 12/20/2022] Open
Abstract
Ghrelin, an orexigenic peptide synthesized by endocrine cells of the gastric mucosa, is released in the bloodstream in response to a negative energetic status. Since discovery, the hypothalamus was identified as the main source of ghrelin in the CNS, and effects of the peptide have been mainly observed in this area of the brain. In recent years, an increasing number of studies have reported ghrelin synthesis and effects in specific populations of neurons also outside the hypothalamus. Thus, ghrelin activity has been described in midbrain, hindbrain, hippocampus, and spinal cord. The spectrum of functions and biological effects produced by the peptide on central neurons is remarkably wide and complex. It ranges from modulation of membrane excitability, to control of neurotransmitter release, neuronal gene expression, and neuronal survival and proliferation. There is not at present a general consensus concerning the source of ghrelin acting on central neurons. Whereas it is widely accepted that the hypothalamus represents the most important endogenous source of the hormone in CNS, the existence of extra-hypothalamic ghrelin-synthesizing neurons is still controversial. In addition, circulating ghrelin can theoretically be another natural ligand for central ghrelin receptors. This paper gives an overview on the distribution of ghrelin and its receptor across the CNS and critically analyses the data available so far as regarding the effects of ghrelin on central neurotransmission.
Collapse
Affiliation(s)
- F Ferrini
- Dipartimento di Morfofisiologia Veterinaria, Università di Torino, Via Leonardo da Vinci 44, 10095, Grugliasco, Italy
| | | | | | | |
Collapse
|
47
|
Metabolic and cardiovascular effects of ghrelin. INTERNATIONAL JOURNAL OF PEPTIDES 2010; 2010. [PMID: 20798901 PMCID: PMC2925368 DOI: 10.1155/2010/864342] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Accepted: 01/16/2010] [Indexed: 01/28/2023]
Abstract
Ghrelin, an endogenous ligand for the growth hormone secretagogue receptor, is synthesized as a preprohormone and then proteolytically processed to yield a 28-amino acid peptide. This peptide was originally reported to induce growth hormone release; large evidence, however, has indicated many other physiological activities of ghrelin, including regulation of food intake and energy balance, as well as of lipid and glucose metabolism. Ghrelin receptors have been detected in the hypothalamus and the pituitary, but also in the cardiovascular system, where ghrelin exerts beneficial hemodynamic activities. Ghrelin administration acutely improves endothelial dysfunction by increasing nitric oxide bioavailability and normalizes the altered balance between endothelin-1 and nitric oxide within the vasculature of patients with metabolic syndrome. Other cardiovascular effects of ghrelin include improvement of left ventricular contractility and cardiac output, as well as reduction of arterial pressure and systemic vascular resistance. In addition, antinflammatory and antiapoptotic actions of ghrelin have been reported both in vivo and in vitro. This review summarizes the most recent findings on the metabolic and cardiovascular effects of ghrelin through GH-dependent and -independent mechanisms and the possible role of ghrelin as a therapeutic molecule for treating cardiovascular diseases.
Collapse
|
48
|
Effect of ghrelin on glucose-insulin homeostasis: therapeutic implications. INTERNATIONAL JOURNAL OF PEPTIDES 2010; 2010. [PMID: 20700401 PMCID: PMC2911604 DOI: 10.1155/2010/234709] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 11/23/2009] [Indexed: 12/17/2022]
Abstract
Ghrelin is a 28-amino-acid peptide that displays a strong growth hormone- (GH-) releasing activity through the activation of the growth hormone secretagogue receptor (GHSR). The first studies about role of ghrelin were focused on its orexigenic ability, but despite indisputable pharmacological data, the evidence for a physiological role for ghrelin in the control of appetite is much less clear. Mice with targeted deletion of either ghrelin or the GHSR exhibit an essentially normal metabolic phenotype when fed a regular chow diet, suggesting that ghrelin may have a redundant role in the regulation of food intake. RNAs for ghrelin as well as GHSR are expressed in the pancreas of rats and humans and several studies propose that ghrelin could have an important function in glucose homeostasis and insulin release, independent of GH secretion. Low plasma ghrelin levels are associated with elevated fasting insulin levels and insulin resistance, suggesting both physiological and pathophysiological roles for ghrelin. For this reason, at least theoretically, ghrelin and/or its signalling manipulation could be useful for the treatment or prevention of diseases of glucose homeostasis such as type 2 diabetes.
Collapse
|
49
|
Stengel A, Goebel M, Wang L, Taché Y. Ghrelin, des-acyl ghrelin and nesfatin-1 in gastric X/A-like cells: role as regulators of food intake and body weight. Peptides 2010; 31:357-69. [PMID: 19944123 PMCID: PMC3166546 DOI: 10.1016/j.peptides.2009.11.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Revised: 11/12/2009] [Accepted: 11/16/2009] [Indexed: 02/07/2023]
Abstract
Numerous peptides released from endocrine cells in the intestinal mucosa were established early on to be involved in the physiological regulation of food intake with a prominent role in termination of food ingestion when nutrients pass along the intestinal tract. Recently, peptides released from X/A-like endocrine cells of the gastric oxyntic mucosa were recognized as additional key players in the regulation of feeding and energy expenditure. Gastric X/A-like cells release the octanoylated peptide, ghrelin, the only known peripherally produced hormone stimulating food intake through interaction with growth hormone secretagogue 1a receptor (GHS-R1a). Additionally, non-octanoylated (des-acyl) ghrelin present in the circulation at higher levels than ghrelin is currently discussed as potential modulator of food intake by opposing ghrelin's action independent from GHS-R1a although the functional significance remains to be established. Obestatin, a ghrelin-associated peptide was initially reported as anorexigenic modulator of ghrelin's orexigenic action. However, subsequent reports did not support this contention. Interesting is the recent identification of nesfatin-1, a peptide derived from the nucleobindin2 gene prominently expressed in gastric X/A-like cells in different vesicles than ghrelin. Circulating nesfatin-1 levels vary with metabolic state and peripheral or central injection inhibits dark phase feeding in rodents. Overall, these data point to an important role of gastric X/A-like cells in food intake regulation through the expression of the orexigenic peptide ghrelin along with des-acyl ghrelin and nesfatin-1 capable of reducing food intake upon exogenous injection although their mechanisms of action and functional significance remain to be established.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE: Digestive Diseases Research Center and Neurobiology of Stress, Digestive Diseases Division, David Geffen School of Medicine at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| | | | | | | |
Collapse
|
50
|
Interactions of gastrointestinal peptides: ghrelin and its anorexigenic antagonists. INTERNATIONAL JOURNAL OF PEPTIDES 2010; 2010. [PMID: 20798884 PMCID: PMC2925274 DOI: 10.1155/2010/817457] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 10/13/2009] [Accepted: 10/19/2009] [Indexed: 12/21/2022]
Abstract
Food intake behaviour and energy homeostasis are strongly regulated by a complex system of humoral factors and nerval structures constituting the brain-gut-axis. To date the only known peripherally produced and centrally acting peptide that stimulates food intake is ghrelin, which is mainly synthesized in the stomach. Recent data indicate that the orexigenic effect of ghrelin might be influenced by other gastrointestinal peptides such as cholecystokinin (CCK), bombesin, desacyl ghrelin, peptide YY (PYY), as well as glucagon-like peptide (GLP). Therefore, we will review on the interactions of ghrelin with several gastrointestinal factors known to be involved in appetite regulation in order to elucidate the interdependency of peripheral orexigenic and anorexigenic peptides in the control of appetite.
Collapse
|