1
|
Farhadi Z, Khaksari M, Alivirdiloo V, Mehrjerdi FZ, Alborzi N, Baktash KB, Rezvani ME. Review on the role of hypothalamic astrocytes in the neuroendocrine control of metabolism. J Diabetes Metab Disord 2024; 23:1635-1643. [PMID: 39610541 PMCID: PMC11599663 DOI: 10.1007/s40200-024-01465-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 11/30/2024]
Abstract
Astrocytes are the most numerous type of glial cells found in the nervous system. They regulate energy homeostasis in collaboration with the neuronal circuits involved in energy balance. These glial cells are equipped with sensors and receptors for nutrients and metabolic hormones in order to control energy homeostasis. Astrocytes, like hypothalamic appetite-regulating neurons, are vulnerable to the negative consequences of a high-fat diet (HFD) feeding, which is associated with an inflammatory response and transforms them into a reactive astrocyte state, consequently leading to the disruption of energy balance. Additionally, these cells have sexually dimorphic characteristics, which will lead to different metabolic outcomes in males and females. In this review, we will discuss the various physiological and pathophysiological roles of astrocytes in regulating energy balance. Finally, we will discuss the sexual dimorphism in astrocytes and the impact of estrogen on eliciting distinct responses.
Collapse
Affiliation(s)
- Zeinab Farhadi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Khaksari
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Vahid Alivirdiloo
- Medical Doctor Ramsar Campus, Mazandaran University of Medical Sciences, Ramsar, Iran
| | - Fatemeh Zare Mehrjerdi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Nasrin Alborzi
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | | | - Mohammad Ebrahim Rezvani
- Yazd Neuroendocrine Research Center, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| |
Collapse
|
2
|
Azuma M, Konno N, Sakata I, Koshimizu TA, Kaiya H. Molecular characterization and distribution of motilin and motilin receptor in the Japanese medaka Oryzias latipes. Cell Tissue Res 2024; 397:61-76. [PMID: 38727755 DOI: 10.1007/s00441-024-03896-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/30/2024] [Indexed: 07/09/2024]
Abstract
Motilin (MLN) is a peptide hormone originally isolated from the mucosa of the porcine intestine. Its orthologs have been identified in various vertebrates. Although MLN regulates gastrointestinal motility in tetrapods from amphibians to mammals, recent studies indicate that MLN is not involved in the regulation of isolated intestinal motility in zebrafish, at least in vitro. To determine the unknown function of MLN in teleosts, we examined the expression of MLN and the MLN receptor (MLNR) at the cellular level in Japanese medaka (Oryzias latipes). Quantitative PCR revealed that mln mRNA was limitedly expressed in the gut, whereas mlnr mRNA was not detected in the gut but was expressed in the brain and kidney. By in situ hybridization and immunohistochemistry, mlnr mRNA was detected in the dopaminergic neurons of the area postrema in the brain and the noradrenaline-producing cells in the interrenal gland of the kidney. Furthermore, we observed efferent projections of mlnr-expressing dopaminergic neurons in the lobus vagi (XL) and nucleus motorius nervi vagi (NXm) of the medulla oblongata by establishing a transgenic medaka expressing the enhanced green fluorescence protein driven by the mlnr promoter. The expression of dopamine receptor mRNAs in the XL and cholinergic neurons in NXm was confirmed by in situ hybridization. These results indicate novel sites of MLN activity other than the gastrointestinal tract. MLN may exert central and peripheral actions through the regulation of catecholamine release in medaka.
Collapse
Affiliation(s)
- Morio Azuma
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, Japan.
| | - Norifumi Konno
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama, 3190, Japan
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimookubo, Saitama, Japan
| | - Taka-Aki Koshimizu
- Division of Molecular Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, Japan
| | - Hiroyuki Kaiya
- Department of Biological Science, Graduate School of Science and Engineering, University of Toyama, Gofuku, Toyama, 3190, Japan
- Division of Drug Discovery, Grandsoul Research Institute for Immunology, Inc. 8-1 Utano-Matsui, Uda, Nara, Japan
| |
Collapse
|
3
|
Vijayashankar U, Ramashetty R, Rajeshekara M, Vishwanath N, Yadav AK, Prashant A, Lokeshwaraiah R. Leptin and ghrelin dynamics: unraveling their influence on food intake, energy balance, and the pathophysiology of type 2 diabetes mellitus. J Diabetes Metab Disord 2024; 23:427-440. [PMID: 38932792 PMCID: PMC11196531 DOI: 10.1007/s40200-024-01418-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 03/12/2024] [Indexed: 06/28/2024]
Abstract
Purpose Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder characterized by insulin resistance and impaired glucose homeostasis. In recent years, there has been growing interest in the role of hunger and satiety hormones such as ghrelin and leptin in the development and progression of T2DM. In this context, the present literature review aims to provide a comprehensive overview of the current understanding of how ghrelin and leptin influences food intake and maintain energy balance and its implications in the pathophysiology of T2DM. Methods A thorough literature search was performed using PubMed and Google Scholar to choose the studies that associated leptin and ghrelin with T2DM. Original articles and reviews were included, letters to editors and case reports were excluded. Results This narrative review article provides a comprehensive summary on mechanism of action of leptin and ghrelin, its association with obesity and T2DM, how they regulate energy and glucose homeostasis and potential therapeutic implications of leptin and ghrelin in managing T2DM. Conclusion Ghrelin, known for its appetite-stimulating effects, and leptin, a hormone involved in the regulation of energy balance, have been implicated in insulin resistance and glucose metabolism. Understanding the complexities of ghrelin and leptin interactions in the context of T2DM may offer insights into novel therapeutic strategies for this prevalent metabolic disorder. Further research is warranted to elucidate the molecular mechanisms underlying these hormone actions and to explore their clinical implications for T2DM prevention and management.
Collapse
Affiliation(s)
- Uma Vijayashankar
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Rajalakshmi Ramashetty
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Mahesh Rajeshekara
- Department of Surgical Gastroenterology, Bangalore Medical College and Research Institute, Bangalore, 560002 India
| | - Nagashree Vishwanath
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| | - Anshu Kumar Yadav
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru-15, Mysuru, 570015 India
| | - Akila Prashant
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru-15, Mysuru, 570015 India
| | - Rajeshwari Lokeshwaraiah
- Department of Physiology, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru, 570015 India
| |
Collapse
|
4
|
Zhang X, Zeng Z, Liu Y, Liu D. Emerging Relevance of Ghrelin in Programmed Cell Death and Its Application in Diseases. Int J Mol Sci 2023; 24:17254. [PMID: 38139082 PMCID: PMC10743592 DOI: 10.3390/ijms242417254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/29/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Ghrelin, comprising 28 amino acids, was initially discovered as a hormone that promotes growth hormones. The original focus was on the effects of ghrelin on controlling hunger and satiation. As the research further develops, the research scope of ghrelin has expanded to a wide range of systems and diseases. Nevertheless, the specific mechanisms remain incompletely understood. In recent years, substantial studies have demonstrated that ghrelin has anti-inflammatory, antioxidant, antiapoptotic, and other effects, which could affect the signaling pathways of various kinds of programmed cell death (PCD) in treating diseases. However, the regulatory mechanisms underlying the function of ghrelin in different kinds of PCD have not been thoroughly illuminated. This review describes the relationship between ghrelin and four kinds of PCD (apoptosis, necroptosis, autophagy, and pyroptosis) and then introduces the clinical applications based on the different features of ghrelin.
Collapse
Affiliation(s)
- Xue Zhang
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Zihan Zeng
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Yaning Liu
- Queen Mary College, Nanchang University, Xuefu Road, Nanchang 330001, China; (X.Z.); (Z.Z.); (Y.L.)
| | - Dan Liu
- School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
5
|
Ma Y, Zhang H, Guo W, Yu L. Potential role of ghrelin in the regulation of inflammation. FASEB J 2022; 36:e22508. [PMID: 35983825 DOI: 10.1096/fj.202200634r] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/27/2022] [Accepted: 08/08/2022] [Indexed: 11/11/2022]
Abstract
Several diseases are caused or progress due to inflammation. In the past few years, accumulating evidence suggests that ghrelin, a gastric hormone of 28-amino acid residue length, exerts protective effects against inflammation by modulating the related pathways. This review focuses on ghrelin's anti-inflammatory and potential therapeutic effects in neurological, cardiovascular, respiratory, hepatic, gastrointestinal, and kidney disorders. Ghrelin significantly alleviates excessive inflammation and reduces damage to different target organs mainly by reducing the secretion of inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α), and inhibiting the nuclear factor kappa-B (NF-κB) and NLRP3 inflammasome signaling pathways. Ghrelin also regulates inflammation and apoptosis through the p38 MAPK/c-Jun N-terminal kinase (JNK) signaling pathway; restores cerebral microvascular integrity, and attenuates vascular leakage. Ghrelin activates the phosphoInositide-3 kinase (PI3K)/protein kinase B (Akt) pathway and inhibits inflammatory responses in cardiovascular diseases and acute kidney injury. Some studies show that ghrelin exacerbates colonic and intestinal manifestations of colitis. Interestingly, some inflammatory states, such as non-alcoholic steatohepatitis, inflammatory bowel diseases, and chronic kidney disease, are often associated with high ghrelin levels. Thus, ghrelin may be a potential new therapeutic target for inflammation-related diseases.
Collapse
Affiliation(s)
- Yunxiao Ma
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haifeng Zhang
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Weiying Guo
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Lu Yu
- Department of Endocrinology and Department of Interventional Therapy of First Hospital of Jilin University, State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
6
|
Binding domain characterization of growth hormone secretagogue receptor. J Transl Int Med 2022; 10:146-155. [PMID: 35959447 PMCID: PMC9328036 DOI: 10.2478/jtim-2022-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background and Objectives Activation of ghrelin receptor growth hormone secretagogue receptor (GHS-R) by endogenous or synthetic ligands amplifies pulsatile release of growth hormone (GH) and enhances food intake, very relevant to development and growth. GHS-R is a G-protein coupled receptor that has great druggable potential. Understanding the precise ligand and receptor interactions is crucial to advance the application of GHS-R. Materials and Methods We used radiolabeled ligand-binding assay and growth hormone release assay to assess the binding and functional characteristics of GHS-R to synthetic agonists MK-0677 and GHS-25, as well as to endogenous peptide ligand ghrelin. We analyzed the ligand-dependent activity of GHS-R by measuring aequorin-based [Ca++]i responses. To define a ligand-binding pocket of GHS-R, we generated a series of human/puffer fish GHS-R chimeras by domain swapping, as well as a series of mutants by site-directed mutagenesis. Results We found that the synthetic ligands have high binding affinity to GHS-R in the in vitro competitive binding assay. Remarkably, the in vivo GH secretagogue activity is higher with the synthetic agonists MK-0677 and GHS-25 than that of ghrelin. Importantly, the activity was completely abolished in GHS-R knockout mice. In GHS-R chimera analysis, we identified the C-terminal region, particularly the transmembrane domain 6 (TM6), to be critical for the ligand-dependent activity. Our site-directed mutagenesis study further revealed that amino acid residues D99 and W276 in GHS-R are essential for ligand binding. Interestingly, critical residues distinctively interact with different ligands, MK-0677 activation depends on E124, while ghrelin and GHS-25 preferentially interact with F279. Conclusion The ligand-binding pocket of human GHS-R is mainly defined by interactive residues in TM6 and the adjacent region of the receptor. This novel finding in GHS-R binding domains advances the structural/ functional understanding of GHS-R, which will help to select/design better GHS-R agonists/ antagonists for future therapeutic applications.
Collapse
|
7
|
Parikh S, Parikh R, Michael K, Bikovski L, Barnabas G, Mardamshina M, Hemi R, Manich P, Goldstein N, Malcov-Brog H, Ben-Dov T, Glaich O, Liber D, Bornstein Y, Goltseker K, Ben-Bezalel R, Pavlovsky M, Golan T, Spitzer L, Matz H, Gonen P, Percik R, Leibou L, Perluk T, Ast G, Frand J, Brenner R, Ziv T, Khaled M, Ben-Eliyahu S, Barak S, Karnieli-Miller O, Levin E, Gepner Y, Weiss R, Pfluger P, Weller A, Levy C. Food-seeking behavior is triggered by skin ultraviolet exposure in males. Nat Metab 2022; 4:883-900. [PMID: 35817855 PMCID: PMC9314261 DOI: 10.1038/s42255-022-00587-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 05/16/2022] [Indexed: 01/03/2023]
Abstract
Sexual dimorphisms are responsible for profound metabolic differences in health and behavior. Whether males and females react differently to environmental cues, such as solar ultraviolet (UV) exposure, is unknown. Here we show that solar exposure induces food-seeking behavior, food intake, and food-seeking behavior and food intake in men, but not in women, through epidemiological evidence of approximately 3,000 individuals throughout the year. In mice, UVB exposure leads to increased food-seeking behavior, food intake and weight gain, with a sexual dimorphism towards males. In both mice and human males, increased appetite is correlated with elevated levels of circulating ghrelin. Specifically, UVB irradiation leads to p53 transcriptional activation of ghrelin in skin adipocytes, while a conditional p53-knockout in mice abolishes UVB-induced ghrelin expression and food-seeking behavior. In females, estrogen interferes with the p53-chromatin interaction on the ghrelin promoter, thus blocking ghrelin and food-seeking behavior in response to UVB exposure. These results identify the skin as a major mediator of energy homeostasis and may lead to therapeutic opportunities for sex-based treatments of endocrine-related diseases.
Collapse
Affiliation(s)
- Shivang Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roma Parikh
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren Michael
- Department of Human Services, The Max Stern Yezreel Valley Academic College, Yezreel Valley, Israel
| | - Lior Bikovski
- The Myers Neuro-Behavioral Core Facility, Tel Aviv University, Tel Aviv, Israel
- School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| | - Georgina Barnabas
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mariya Mardamshina
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rina Hemi
- Endocrine Service Unit, Sheba Medical Center Hospital, Tel Hashomer, Ramat Gan, Israel
| | - Paulee Manich
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Goldstein
- School of Public Health, Sackler Faculty of Medicine and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Hagar Malcov-Brog
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tom Ben-Dov
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Otolaryngology, Head and Neck surgery, Meir Medical Center, Kfar Saba, Israel
| | - Ohad Glaich
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Daphna Liber
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yael Bornstein
- School of Public Health, Sackler Faculty of Medicine and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Koral Goltseker
- Zuckerman Mind Brain Behavior Institute, Howard Hughes Medical Institute and Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Roy Ben-Bezalel
- School of Zoology, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Mor Pavlovsky
- Division of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Tamar Golan
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liron Spitzer
- Division of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
| | - Hagit Matz
- Division of Dermatology, Tel Aviv Sourasky (Ichilov) Medical Center, Tel Aviv, Israel
- Phototherapy Unit, Assuta Medical Center, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pinchas Gonen
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth Percik
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Division of Endocrinology, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Lior Leibou
- Department of Plastic and Reconstructive Surgery, E. Wolfson Medical Center, Holon, Israel
| | - Tomer Perluk
- Department of Plastic and Reconstructive Surgery, E. Wolfson Medical Center, Holon, Israel
| | - Gil Ast
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Frand
- Department of Plastic and Reconstructive Surgery, E. Wolfson Medical Center, Holon, Israel
| | - Ronen Brenner
- Institute of Oncology, E. Wolfson Medical Center, Holon, Israel
| | - Tamar Ziv
- The Smoler Proteomics Center, Lorry I. Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion, Haifa, Israel
| | - Mehdi Khaled
- INSERM 1279, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Shamgar Ben-Eliyahu
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Segev Barak
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Orit Karnieli-Miller
- Department of Medical Education, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Levin
- School of Zoology, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel
| | - Yftach Gepner
- School of Public Health, Sackler Faculty of Medicine and Sylvan Adams Sports Institute, Tel Aviv University, Tel Aviv, Israel
| | - Ram Weiss
- Department of Pediatrics, Ruth Rappaport Children's Hospital, Rambam Medical Center and Technion School of Medicine, Haifa, Israel
| | - Paul Pfluger
- Research Unit Neurobiology of Diabetes, Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Centre for Diabetes Research (DZD), Neuherberg, Germany
| | - Aron Weller
- Department of Psychology and the Gonda Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| | - Carmit Levy
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
8
|
Vlug LE, Delhanty PJD, Neelis EG, Huisman M, Visser JA, Rings EHHM, Wijnen RMH, Nagelkerke SCJ, Tabbers MM, Hulst JM, de Koning BAE. Ghrelin Levels in Children With Intestinal Failure Receiving Long-Term Parenteral Nutrition. Front Nutr 2022; 9:896328. [PMID: 35634374 PMCID: PMC9131070 DOI: 10.3389/fnut.2022.896328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
Background Children with intestinal failure (IF) require parenteral nutrition (PN). Transition to oral and enteral nutrition (EN) can be difficult also due to abnormal gastrointestinal motility. The gut hormone ghrelin is increased in states of negative energy balance, functioning to preserve euglycemia, and also has appetite stimulating and prokinetic properties. We aimed to evaluate and compare ghrelin levels in children with IF, and to assess the relationship with PN-dependency. Methods In this exploratory prospective multicenter study, plasma acylated (AG) and unacylated (UAG) ghrelin levels were measured in children with short bowel syndrome (SBS) and with functional IF (pseudo-obstruction or any enteropathy) and compared with healthy control subjects. Spearman’s rho (rs) was used to assess correlations of AG and UAG with PN-dependency (%PN) and parenteral glucose intake. Results Sixty-four samples from 36 IF-patients were analyzed. Median baseline AG and UAG levels were respectively 279.2 and 101.0 pg/mL in children with SBS (n = 16), 126.4 and 84.5 pg/mL in children with functional IF (n = 20) and 82.4 and 157.3 pg/mL in healthy children (n = 39). AG levels were higher in children with SBS and functional IF than in healthy children (p = 0.002 and p = 0.023, respectively). In SBS, AG positively correlated with %PN (rs = 0.5, p = 0.005) and parenteral glucose intake (rs = 0.6, p = 0.003). These correlations were not observed in functional IF. Conclusion Children with IF had raised AG levels which could be related to starvation of the gut. The positive correlation between AG and glucose infusion rate in SBS suggests an altered glucoregulatory function.
Collapse
Affiliation(s)
- Lotte E. Vlug
- Division of Gastroenterology, Department of Pediatrics, Erasmus MC Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Patric J. D. Delhanty
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Esther G. Neelis
- Division of Gastroenterology, Department of Pediatrics, Erasmus MC Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martin Huisman
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Edmond H. H. M. Rings
- Division of Gastroenterology, Department of Pediatrics, Erasmus MC Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- Division of Gastroenterology, Department of Pediatrics, Willem Alexander Children’s Hospital, Leiden University Medical Center, Leiden, Netherlands
| | - René M. H. Wijnen
- Department of Pediatric Surgery, Erasmus MC Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sjoerd C. J. Nagelkerke
- Division of Gastroenterology, Department of Pediatrics, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Merit M. Tabbers
- Division of Gastroenterology, Department of Pediatrics, Emma Children’s Hospital, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Jessie M. Hulst
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, ON, Canada
| | - Barbara A. E. de Koning
- Division of Gastroenterology, Department of Pediatrics, Erasmus MC Sophia Children’s Hospital, University Medical Center Rotterdam, Rotterdam, Netherlands
- *Correspondence: Barbara A. E. de Koning,
| |
Collapse
|
9
|
Iacob SA, Iacob DG. Non-Alcoholic Fatty Liver Disease in HIV/HBV Patients - a Metabolic Imbalance Aggravated by Antiretroviral Therapy and Perpetuated by the Hepatokine/Adipokine Axis Breakdown. Front Endocrinol (Lausanne) 2022; 13:814209. [PMID: 35355551 PMCID: PMC8959898 DOI: 10.3389/fendo.2022.814209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with the metabolic syndrome and is one of the most prevalent comorbidities in HIV and HBV infected patients. HIV plays an early and direct role in the development of metabolic syndrome by disrupting the mechanism of adipogenesis and synthesis of adipokines. Adipokines, molecules that regulate the lipid metabolism, also contribute to the progression of NAFLD either directly or via hepatic organokines (hepatokines). Most hepatokines play a direct role in lipid homeostasis and liver inflammation but their role in the evolution of NAFLD is not well defined. The role of HBV in the pathogenesis of NAFLD is controversial. HBV has been previously associated with a decreased level of triglycerides and with a protective role against the development of steatosis and metabolic syndrome. At the same time HBV displays a high fibrogenetic and oncogenetic potential. In the HIV/HBV co-infection, the metabolic changes are initiated by mitochondrial dysfunction as well as by the fatty overload of the liver, two interconnected mechanisms. The evolution of NAFLD is further perpetuated by the inflammatory response to these viral agents and by the variable toxicity of the antiretroviral therapy. The current article discusses the pathogenic changes and the contribution of the hepatokine/adipokine axis in the development of NAFLD as well as the implications of HIV and HBV infection in the breakdown of the hepatokine/adipokine axis and NAFLD progression.
Collapse
Affiliation(s)
- Simona Alexandra Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Infectious Diseases, National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, Bucharest, Romania
| | - Diana Gabriela Iacob
- Department of Infectious Diseases, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
- Department of Infectious Diseases, Emergency University Hospital, Bucharest, Romania
- *Correspondence: Diana Gabriela Iacob,
| |
Collapse
|
10
|
Masule MV, Rathod S, Agrawal Y, Patil CR, Nakhate KT, Ojha S, Goyal SN, Mahajan UB. Ghrelin mediated regulation of neurosynaptic transmitters in depressive disorders. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2022; 3:100113. [PMID: 35782191 PMCID: PMC9240712 DOI: 10.1016/j.crphar.2022.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/07/2022] [Accepted: 06/07/2022] [Indexed: 11/16/2022] Open
Abstract
Ghrelin is a peptide released by the endocrine cells of the stomach and the neurons in the arcuate nucleus of the hypothalamus. It modulates both peripheral and central functions. Although ghrelin has emerged as a potent stimulator of growth hormone release and as an orexigenic neuropeptide, the wealth of literature suggests its involvement in the pathophysiology of affective disorders including depression. Ghrelin exhibits a dual role through the advancement and reduction of depressive behavior with nervousness in the experimental animals. It modulates depression-related signals by forming neuronal networks with various neuropeptides and classical neurotransmitter systems. The present review emphasizes the integration and signaling of ghrelin with other neuromodulatory systems concerning depressive disorders. The role of ghrelin in the regulation of neurosynaptic transmission and depressive illnesses implies that the ghrelin system modulation can yield promising antidepressive therapies. Ghrelin is the orexigenic type of neuropeptide. It binds with the growth hormone secretagogue receptor (GHSR). GHSR is ubiquitously present in the various brain regions. Ghrelin is involved in the regulation of depression-related behavior. The review focuses on the neurotransmission and signaling of ghrelin in neuropsychiatric and depressive disorders.
Collapse
Affiliation(s)
- Milind V. Masule
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Sumit Rathod
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Yogeeta Agrawal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Chandragouda R. Patil
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
| | - Kartik T. Nakhate
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, 424001, Maharashtra, India
- Corresponding author.
| | - Umesh B. Mahajan
- Department of Pharmacology, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, 425405, Maharashtra, India
- Corresponding author.
| |
Collapse
|
11
|
Samimi AS, Aghamiri SM, Nazifi S, Asadi Z. Changes in ghrelin, microminerals, antioxidants and vitamins A, E and C levels during different physiological status in high yielding Saanen goats subjected to heat stress. J Therm Biol 2021; 100:103014. [PMID: 34503772 DOI: 10.1016/j.jtherbio.2021.103014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/25/2021] [Accepted: 05/30/2021] [Indexed: 11/25/2022]
Abstract
The aim of this study was to evaluate the changes of ghrelin, microminerals, antioxidants, and vitamins A, E and C levels during different metabolic periods in high yielding Saanen goats subjected to heat stress. Twenty clinically and paraclinically healthy, high yielding and multiparous goats with an average age of 3 ± 0.5 years and pregnant with a single fetus were included in this study. Sampling was performed at three different physiologic periods: non-pregnancy non-lactation (P1), four-month gestation (P2), and first month of lactation (P3). In this study, the ambient temperature ranged from 19 to 42 °C and relative humidity ranged from 14 to 19% during the hot months. Serum concentrations of ghrelin, glucose (Glu), superoxide dismutase (SOD), glutathione peroxidase (GPx), vitamins (A, E and C) and microminerals (selenium, manganese, cobalt, iron, copper and zinc) were measured. Mean raw milk yield of the goats per day at the first month of lactation was 2.34 ± 0.2 kg. Concentration of ghrelin at P1 was significantly lower than P2 and P3 (P < 0.05). Glucose levels were significantly lower at P3 compared with P1 and P2 (P < 0.05). There were no significant correlations between ghrelin and Glu at different periods. Concentrations of selenium and manganese were significantly higher at P3 compared with P2 and were significantly higher at P2 compared with P1. Values of copper at P2 were significantly higher than P1 and P3 (P < 0.05). Zinc levels were significantly higher at P1 compared with P2 and P3 (P < 0.05). Values of antioxidants and vitamins were significantly lower at P3 compared with P2. It is concluded that high yielding Saanen goats may suffer from hormonal and metabolic disturbances, oxidative stress and micromineral deficiencies during late gestation and the first month of lactation especially when they are subjected to heat stress.
Collapse
Affiliation(s)
- Amir Saeed Samimi
- Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | | | - Saeed Nazifi
- Faculty of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Zahra Asadi
- Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
12
|
Abstract
Obesity is as a global health problem due to its interaction with complex chronic disorders such as cardiovascular disorders, type 2 diabetes mellitus (T2DM) and cancer. Despite the fact that pathogenesis of obesity is not yet clearly understood, it is associated with a combination of psychological, environmental and various genetic factors. Here, employing a case-control design, we aimed to examine the effects of the GHRL c.152C>T (p.Arg51Gln) (rs34911341) and c.214G>T (p.Leu72Met) (rs696217) markers on susceptibility to obesity in a Turkish-Cypriot population, as well as to evaluate whether these markers affect biochemical parameters and show their putative functional consequences. This study involved 211 Turkish-Cypriot subjects (106 obese and 95 non obese). Genotyping for the GHRL gene polymorphisms was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. Our results indicate that the GHRL Leu72Met polymorphism was found to be significantly higher in obese patients, with respect to genotypic (p = 0.0012) and allelic (p = 0.0005) frequencies. Strikingly, the rs696217 GT genotype (heterozygous) had significantly lower serum high-density lipoprotein cholesterol (HDL-C) (p = 0.015) than GG (wild type) genotypes. Overall, Leu72Met susceptibility variant may be considered as risk and crucial marker for both obesity and cholesterol metabolism in the community of Turkish-Cypriots. Thus, the dual effect of the GHRL gene Leu72Met variant may be used for clinical diagnosis.
Collapse
|
13
|
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: 1.3] [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
|
14
|
Tolle V, Ramoz N, Epelbaum J. Is there a hypothalamic basis for anorexia nervosa? HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:405-424. [PMID: 34238474 DOI: 10.1016/b978-0-12-820683-6.00030-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The hypothalamus has long been known to control food intake and energy metabolism through a complex network of primary and secondary neurons and glial cells. Anorexia nervosa being a complex disorder characterized by abnormal feeding behavior and food aversion, it is thus quite surprising that not much is known concerning potential hypothalamic modifications in this disorder. In this chapter, we review the recent advances in the fields of genetics, epigenetics, structural and functional imaging, and brain connectivity, as well as neuroendocrine findings and emerging animal models, which have begun to unravel the importance of hypothalamic adaptive processes to our understanding of the pathology of eating disorders.
Collapse
|
15
|
Barja-Fernández S, Lugilde J, Castelao C, Vázquez-Cobela R, Seoane LM, Diéguez C, Leis R, Tovar S. Circulating LEAP-2 is associated with puberty in girls. Int J Obes (Lond) 2020; 45:502-514. [PMID: 33139887 DOI: 10.1038/s41366-020-00703-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 09/25/2020] [Accepted: 10/23/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES Liver-expressed antimicrobial peptide 2 (LEAP-2) was recently identified as an endogenous non-competitive allosteric antagonist of the growth hormone secretagogue receptor 1a (GHSR1a). LEAP-2 blunts ghrelin-induced feeding and its plasma levels are modulated in response to nutritional status in humans. Despite the relevant role of ghrelin in childhood, puberty, and childhood obesity, the potential implication of LEAP-2 in these aspects remains totally unknown. We aimed to investigate the regulation of circulating plasma LEAP-2 in childhood and adolescent either lean or obese. METHODS AND RESULTS Plasma levels of LEAP-2 were analyzed in a cross-sectional study with lean and obese children and adolescents (n = 150). Circulating LEAP-2 levels were significantly higher in girls than in boys independently of whether they were obese or lean. In addition, LEAP-2 was significantly increased (p < 0.001) in pubertal than in prepubertal girls, while no changes were found in boys between both developmental stages. Moreover, in girls LEAP-2 was positively correlated with insulin, IGF-1, HOMA-IR and triglycerides and negatively with ghrelin. In boys, LEAP-2 was positively correlated with leptin and negatively with vitamin D levels. CONCLUSION This study reveals a sexual dimorphism in LEAP-2 levels in children and adolescents. These changes and the higher levels during puberty imply that LEAP-2 may contribute to some of the biological adaptations occurring during pubertal development in terms of food intake, energy balance, growth rate, and puberty onset. Future studies assessing LEAP-2 levels in longitudinal studies and its implications in growth rate, puberty onset, and reproductive hormones will help to understand the relevance of this hormone in this stage of life.
Collapse
Affiliation(s)
- Silvia Barja-Fernández
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), 15706, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain.,Departamento Pediatría, GI Nutrición Pediátrica (IDIS,CHUS), Unidad de investigación de Galicia de desarrollo, crecimiento y nutrición humana. Universidade de Santiago de Compostela (USC), 15706, Santiago de Compostela, Spain
| | - Javier Lugilde
- Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular y Enfermedades Cronicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Santiago de Compostela, 15782, Spain
| | - Cecilia Castelao
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), 15706, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain
| | - Rocío Vázquez-Cobela
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain.,Departamento Pediatría, GI Nutrición Pediátrica (IDIS,CHUS), Unidad de investigación de Galicia de desarrollo, crecimiento y nutrición humana. Universidade de Santiago de Compostela (USC), 15706, Santiago de Compostela, Spain
| | - Luisa M Seoane
- Grupo Fisiopatología Endocrina, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo. Hospitalario Universitario de Santiago (CHUS/SERGAS), 15706, Santiago de Compostela, Spain.,CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain
| | - Carlos Diéguez
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain.,Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular y Enfermedades Cronicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Santiago de Compostela, 15782, Spain
| | - Rosaura Leis
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain. .,Departamento Pediatría, GI Nutrición Pediátrica (IDIS,CHUS), Unidad de investigación de Galicia de desarrollo, crecimiento y nutrición humana. Universidade de Santiago de Compostela (USC), 15706, Santiago de Compostela, Spain.
| | - Sulay Tovar
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Santiago de Compostela, 15706, Spain. .,Departamento de Fisioloxía and Centro de Investigación en Medicina Molecular y Enfermedades Cronicas (CIMUS), Universidade de Santiago de Compostela, Instituto de Investigaciones Sanitarias de Santiago de Compostela (IDIS), Santiago de Compostela, 15782, Spain.
| |
Collapse
|
16
|
González‐López MA, Ocejo‐Viñals JG, Mata C, Vilanova I, Guiral S, Portilla V, Blanco R, Hernández JL. Association of retinol binding protein4 (RBP4) and ghrelin plasma levels with insulin resistance and disease severity in non‐diabetic patients with hidradenitis suppurativa. Exp Dermatol 2020; 29:828-832. [DOI: 10.1111/exd.14132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/26/2020] [Accepted: 06/14/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Marcos A. González‐López
- Division of Dermatology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - J. Gonzalo Ocejo‐Viñals
- Division of Immunology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - Cristina Mata
- Division of Rheumatology Hospital Comarcal Laredo Spain
| | - Iosune Vilanova
- Division of Dermatology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - Sandra Guiral
- Division of Immunology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - Virginia Portilla
- Division of Rheumatology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - Ricardo Blanco
- Division of Rheumatology Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| | - José L. Hernández
- Division of Internal Medicine Hospital Universitario Marqués de Valdecilla University of CantabriaIDIVAL Santander Spain
| |
Collapse
|
17
|
Moose JE, Leets KA, Mate NA, Chisholm JD, Hougland JL. An overview of ghrelin O-acyltransferase inhibitors: a literature and patent review for 2010-2019. Expert Opin Ther Pat 2020; 30:581-593. [PMID: 32564644 DOI: 10.1080/13543776.2020.1776263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The peptide hormone ghrelin regulates physiological processes associated with energy homeostasis such as appetite, insulin signaling, glucose metabolism, and adiposity. Ghrelin has also been implicated in a growing number of neurological pathways involved in stress response and addiction behavior. For ghrelin to bind the growth hormone secretagogue receptor 1a (GHS-R1a) and activate signaling, the hormone must first be octanoylated on a specific serine side chain. This key transformation is performed by the enzyme ghrelin O-acyltransferase (GOAT), and therefore GOAT inhibitors may be useful in treating disorders related to ghrelin signaling such as diabetes, obesity, and related metabolic syndromes. AREAS COVERED This report covers ghrelin and GOAT as potential therapeutic targets and summarizes work on GOAT inhibitors through the end of 2019, highlighting recent successes with both peptidomimetics and small molecule GOAT inhibitors as potent modulators of GOAT-catalyzed ghrelin octanoylation. EXPERT OPINION A growing body of biochemical and structural knowledge regarding the ghrelin/GOAT system now enables multiple avenues for identifying and optimizing GOAT inhibitors. We are at the beginning of a new era with increased opportunities for leveraging ghrelin and GOAT in the understanding and treatment of multiple health conditions including diabetes, obesity, and addiction.
Collapse
Affiliation(s)
- Jacob E Moose
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - Katelyn A Leets
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - Nilamber A Mate
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - John D Chisholm
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - James L Hougland
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| |
Collapse
|
18
|
Veiga L, Brito M, Silva C, Silva-Nunes J. Glucose Homeostasis in Obese Women Is Not Associated to Unacylated Ghrelin Plasma Levels. Biomark Insights 2020; 15:1177271920928923. [PMID: 32550765 PMCID: PMC7278305 DOI: 10.1177/1177271920928923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 01/12/2023] Open
Abstract
Introduction: Unacylated ghrelin (UAG) is the major form of circulating ghrelin. Initially considered as a nonfunctional peptide, soon after, UAG has been associated to an insulin sensitizing action and to a negative action on energy balance. The aim of this study was to analyze the association between the serum levels of UAG and glucose metabolism parameters in obese women, independently from eventual influence of anthropometrics. Methods: One hundred lean and 254 obese Caucasian women were studied. Each woman was characterized for anthropometrics, fasting glucose, insulin, HbA1c, and UAG. In addition, obese women were subjected to a classic oral glucose tolerance test (oGTT) to assess glucose and insulin at 120 minutes. Insulin resistance was assessed by the homeostasis model assessment (HOMA-IR). Obese women were classified in 3 glycemic status subgroups (normoglycemia, prediabetes, and diabetes) according to HbA1c and to fasting and oGTT glucose values. Results: In comparison with the lean group, significantly lower levels of UAG were observed in obese women. However, no significant difference was observed through obesity classes I to III. UAG levels were not significantly different among glycemic status subgroups and did not show any association with glucose, insulin, HOMA-IR, or HbA1c. Conclusions: Although anthropometry can influence the level of the unacylated form of ghrelin, UAG plasma levels do not associate to glucose homeostasis parameters.
Collapse
Affiliation(s)
- Luisa Veiga
- Health and Technology Research Center (H&TRC), Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Miguel Brito
- Health and Technology Research Center (H&TRC), Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Carina Silva
- Health and Technology Research Center (H&TRC), Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal.,Centro de Estatística e Aplicações, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - José Silva-Nunes
- Health and Technology Research Center (H&TRC), Escola Superior de Tecnologia da Saúde de Lisboa, Instituto Politécnico de Lisboa, Lisbon, Portugal.,Department of Endocrinology, Diabetes and Metabolism, Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal.,NOVA Medical School-Faculdade de Ciências Medicas, New University of Lisbon, Lisbon, Portugal
| |
Collapse
|
19
|
Ghrelin infused into the dorsomedial hypothalamus of male mice increases food intake and adiposity. Physiol Behav 2020; 220:112882. [PMID: 32205145 DOI: 10.1016/j.physbeh.2020.112882] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a 28 amino acid peptide hormone that targets the brain to promote feeding and adiposity. The ghrelin receptor, the GHSR1a, is expressed within most hypothalamic nuclei, including the DMH, but the role of GHSR1a in this region on energy balance is unknown. In order to investigate whether GHSR1a within the DMH modulate energy balance, we implanted osmotic minipumps filled with saline, ghrelin, or the GHSR1a antagonist JMV2959, and connected it to a cannula aimed unilaterally at the DMH of adult male C57BLJ6 mice and assessed their metabolic profile. We found that chronic infusion of ghrelin in the DMH promoted an increase in caloric intake as well as a decrease in energy expenditure. This translated to an overall increase in weight gain, primarily in the form of adipose tissue in ghrelin treated animals. Further, chronic ghrelin unilateral infusion into the DMH slowed glucose clearance. These results suggest that GHSR in the DMH significantly contribute to the metabolic effects produced by ghrelin.
Collapse
|
20
|
Ilie IR. Neurotransmitter, neuropeptide and gut peptide profile in PCOS-pathways contributing to the pathophysiology, food intake and psychiatric manifestations of PCOS. Adv Clin Chem 2019; 96:85-135. [PMID: 32362321 DOI: 10.1016/bs.acc.2019.11.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Polycystic ovary syndrome (PCOS) is a major health problem with a heterogeneous hormone-imbalance and clinical presentation across the lifespan of women. Increased androgen production and abnormal gonadotropin-releasing hormone (GnRH) release and gonadotropin secretion, resulting in chronic anovulation are well-known features of the PCOS. The brain is both at the top of the neuroendocrine axis regulating ovarian function and a sensitive target of peripheral gonadal hormones and peptides. Current literature illustrates that neurotransmitters regulate various functions of the body, including reproduction, mood and body weight. Neurotransmitter alteration could be one of the reasons for disturbed GnRH release, consequently directing the ovarian dysfunction in PCOS, since there is plenty evidence for altered catecholamine metabolism and brain serotonin or opioid activity described in PCOS. Further, the dysregulated neurotransmitter and neuropeptide profile in PCOS could also be the reason for low self-esteem, anxiety, mood swings and depression or obesity, features closely associated with PCOS women. Can these altered central brain circuits, or the disrupted gut-brain axis be the tie that would both explain and link the pathogenesis of this disorder, the occurrence of depression, anxiety and other mood disorders as well as of obesity, insulin resistance and abnormal appetite in PCOS? This review intends to provide the reader with a comprehensive overview of what is known about the relatively understudied, but very complex role that neurotransmitters, neuropeptides and gut peptides play in PCOS. The answer to the above question may help the development of drugs to specifically target these central and peripheral circuits, thereby providing a valuable treatment for PCOS patients that present to the clinic with GnRH/LH hypersecretion, obesity or psychiatric manifestations.
Collapse
Affiliation(s)
- Ioana R Ilie
- Department of Endocrinology, University of Medicine and Pharmacy 'Iuliu-Hatieganu', Cluj-Napoca, Romania.
| |
Collapse
|
21
|
Magherini F, Fiaschi T, Marzocchini R, Mannelli M, Gamberi T, Modesti PA, Modesti A. Oxidative stress in exercise training: the involvement of inflammation and peripheral signals. Free Radic Res 2019; 53:1155-1165. [PMID: 31762356 DOI: 10.1080/10715762.2019.1697438] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The evidence about the health benefits of regular physical activity is well established. Exercise intensity is a significant variable and structured high-intensity interval training (HIIT) has been demonstrated to improve both whole-body and skeletal muscle metabolic health in different populations. Conversely, fatigue accumulation, if not resolved, leads to overwork, chronic fatigue syndrome (CFS), overtraining syndrome up to alterations of endocrine function, immune, systemic inflammation, and organic diseases with health threat. In response to temporary increases in stress during training, some athletes are unable to maintain sufficient caloric intake, thus suffering a negative energy balance that causes further stress. The regulation of the energy balance is controlled by the central nervous system through an elaborate interaction of the signalling that involves different tissues such as leptin, adiponectin and ghrelin whose provide important feedback to the hypothalamus to regulate the energy balance. Although exercise-induced reactive oxygen species are required for normal force production in muscle, high levels of ROS appear to promote contractile dysfunction. However, a high level of oxidative stress in may induce a rise in inflammatory markers and a disregulation in expression of adiponectin, leptin and grelin.
Collapse
Affiliation(s)
- Francesca Magherini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Tania Fiaschi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Riccardo Marzocchini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Michele Mannelli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Tania Gamberi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pietro Amedeo Modesti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Modesti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
22
|
Su M, Yan M, Gong Y. Ghrelin fiber projections from the hypothalamic arcuate nucleus into the dorsal vagal complex and the regulation of glycolipid metabolism. Neuropeptides 2019; 78:101972. [PMID: 31610887 DOI: 10.1016/j.npep.2019.101972] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/11/2019] [Accepted: 09/14/2019] [Indexed: 12/23/2022]
Abstract
OBJECTIVES This study aimed to explore the involvement of the ghrelin pathway from the arcuate nucleus (ARC) to the dorsal vagal complex (DVC) and to determine its role in the regulation of glycolipid metabolism. METHODS The protein and mRNA expression of ghrelin and growth hormone (GH) secretagogue receptor type 1a (GHSR-1a) were measured using immunohistochemistry and the polymerase chain reaction (PCR) method, respectively. Ghrelin fiber projections arising from the ARC and projecting into the DVC were investigated using retrograde tracing, combined with fluorescence immunohistochemical staining. The effects of electrical stimulation (ES) of the ARC on ghrelin-responsive, glucose-sensitive DVC neurons, glycolipid metabolism, and liver lipid enzymes were determined using electrical physiological method, biochemical analysis, quantitative real-time PCR (qRT-PCR) and Western blot analysis. RESULTS GHSR-1a was expressed in the DVC neurons. Ghrelin fibers originating from the ARC projected into the DVC. ES of the ARC-activated the ghrelin-responsive glucose-excited (GE) and glucose-inhibited (GI) neurons in the DVC. ES of the ARC significantly elevated the serum triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and glucose levels; it reduced the serum high-density lipoprotein (HDLC) and insulin levels. Moreover, ES of the ARC increased liver acetyl-CoA carboxylase-1 (ACC-1) and decreased carnitine palmitoyltransferase-1 (CPT-1) expression, resulting in lipid accumulation in the liver. All the aforementioned effects were partially blocked by pretreatment with the ghrelin receptor antagonist [D-Lys-3]-GHRP-6 in the DVC and were reduced by vagotomy. ES of the ARC increased agouti-related protein (AgRP)/neuropeptide Y (NPY) expression in the ARC and ghrelin expression in the DVC. CONCLUSION Ghrelin fiber projections arising from the ARC and projecting into the DVC play a role in the regulation of afferent glucose metabolism and glycolipid metabolism via the ghrelin receptor GHSR-1a in the DVC.
Collapse
Affiliation(s)
- Manqing Su
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Meixing Yan
- Qingdao Women and Children's Hospital, Qingdao 266042, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
23
|
Affiliation(s)
- Zane B Andrews
- Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
24
|
Mani BK, Shankar K, Zigman JM. Ghrelin's Relationship to Blood Glucose. Endocrinology 2019; 160:1247-1261. [PMID: 30874792 PMCID: PMC6482034 DOI: 10.1210/en.2019-00074] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/09/2019] [Indexed: 12/16/2022]
Abstract
Much effort has been directed at studying the orexigenic actions of administered ghrelin and the potential effects of the endogenous ghrelin system on food intake, food reward, body weight, adiposity, and energy expenditure. Although endogenous ghrelin's actions on some of these processes remain ambiguous, its glucoregulatory actions have emerged as well-recognized features during extreme metabolic conditions. The blood glucose-raising actions of ghrelin are beneficial during starvation-like conditions, defending against life-threatening falls in blood glucose, but they are seemingly detrimental in obese states and in certain monogenic forms of diabetes, contributing to hyperglycemia. Also of interest, blood glucose negatively regulates ghrelin secretion. This article reviews the literature suggesting the existence of a blood glucose-ghrelin axis and highlights the factors that mediate the glucoregulatory actions of ghrelin, especially during metabolic extremes such as starvation and diabetes.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kripa Shankar
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey M Zigman
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
- Correspondence: Jeffrey M. Zigman, MD, PhD, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390. E-mail:
| |
Collapse
|
25
|
Dominguez Gutierrez G, Kim J, Lee AH, Tong J, Niu J, Gray SM, Wei Y, Ding Y, Ni M, Adler C, Murphy AJ, Gromada J, Xin Y. Gene Signature of the Human Pancreatic ε Cell. Endocrinology 2018; 159:4023-4032. [PMID: 30380031 PMCID: PMC6963699 DOI: 10.1210/en.2018-00833] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 10/17/2018] [Indexed: 12/31/2022]
Abstract
The ghrelin-producing ε cell represents the fifth endocrine cell type in human pancreatic islets. The abundance of ε cells in adult pancreas is extremely low, which has hampered the investigation on the molecular pathways regulating the development and the function of this cell type. In this study, we explored the molecular features defining the function of pancreatic ε cells isolated from adult nondiabetic donors using single-cell RNA sequencing technology. We focus on transcription factors, cell surface receptors, and genes involved in metabolic pathways that contribute to regulation of cellular function. Furthermore, the genes that separate ε cells from the other islet endocrine cell types are presented. This study expands prior knowledge about the genes important for ε cell functioning during development and provides a resource to interrogate the transcriptome of this rare human islet cell type.
Collapse
Affiliation(s)
| | - Jinrang Kim
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Ann-Hwee Lee
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Jenny Tong
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - JingJing Niu
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Sarah M Gray
- Division of Endocrinology, Metabolism and Nutrition, Duke Molecular Physiology Institute, Duke University, Durham, North Carolina
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Yueming Ding
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| | | | | | | | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
26
|
Fasting and postprandial acyl and desacyl ghrelin and the acyl/desacyl ratio in obese patients before and after different types of bariatric surgery. Wideochir Inne Tech Maloinwazyjne 2018; 13:366-375. [PMID: 30302150 PMCID: PMC6174170 DOI: 10.5114/wiitm.2018.75868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/04/2018] [Indexed: 12/19/2022] Open
Abstract
Introduction The mechanism underlying beneficial outcomes of bariatric surgery still remains unclear. Especially little is known about hormonal and metabolic changes induced by the novel bariatric procedure mini gastric bypass (MGB). Aim To evaluate pre- and post-prandial changes in both ghrelin isoforms in obese patients without diabetes and cardiovascular complications treated with MGB, sleeve gastrectomy (SG) or Roux-en-Y gastric bypass (RYGB) surgery. Material and methods From 45 patients initially enrolled in the study, 23 persons completed a one-year follow-up period. Venous blood for acyl and desacyl ghrelin (AG and DAG) as well as other metabolic assays was collected 3 months before and 6 and 12 months after bariatric surgery (MGB, RYGB, SG) – in the fasting state and 2 h after the consumption of a standard 300 kcal-mixed meal (Nutridrink standard, Nutricia). Results AG and DAG levels (both fasting and prandial) as well as AG/DAG ratio did not change after 6 and 12 months in MGB and RYGB groups. In the SG group we observed a significant decrease in fasting and postprandial DAG levels and consecutively an increase in the fasting AG/DAG ratio after 6 and 12 months. Six months after surgery we observed some differences between carbohydrate metabolism measures in the MGB group (lower HbA1c, HOMA-IR and fasting insulinaemia) in comparison to the rest of the participants, but 12 months after each type of surgery body mass index and indices of carbohydrate and lipid metabolism did not differ. Conclusions The results of our study demonstrate that all studied bariatric procedures can successfully reduce overall body weight and suggest also that the mechanisms of weight loss and improvement in carbohydrate and lipid metabolism after all three types of surgery are independent of ghrelin and the acyl/desacyl ghrelin ratio.
Collapse
|
27
|
Cleverdon ER, Davis TR, Hougland JL. Functional group and stereochemical requirements for substrate binding by ghrelin O-acyltransferase revealed by unnatural amino acid incorporation. Bioorg Chem 2018; 79:98-106. [PMID: 29738973 DOI: 10.1016/j.bioorg.2018.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/02/2018] [Accepted: 04/13/2018] [Indexed: 12/22/2022]
Abstract
Ghrelin is a small peptide hormone that undergoes a unique posttranslational modification, serine octanoylation, to play its physiological roles in processes including hunger signaling and glucose metabolism. Ghrelin O-acyltransferase (GOAT) catalyzes this posttranslational modification, which is essential for ghrelin to bind and activate its cognate GHS-R1a receptor. Inhibition of GOAT offers a potential avenue for modulating ghrelin signaling for therapeutic effect. Defining the molecular characteristics of ghrelin that lead to binding and recognition by GOAT will facilitate the development and optimization of GOAT inhibitors. We show that small peptide mimics of ghrelin substituted with 2,3-diaminopropanoic acid in place of the serine at the site of octanoylation act as submicromolar inhibitors of GOAT. Using these chemically modified analogs of desacyl ghrelin, we define key functional groups within the N-terminal sequence of ghrelin essential for binding to GOAT and determine GOAT's tolerance to backbone methylations and altered amino acid stereochemistry within ghrelin. Our study provides a structure-activity analysis of ghrelin binding to GOAT that expands upon activity-based investigations of ghrelin recognition and establishes a new class of potent substrate-mimetic GOAT inhibitors for further investigation and therapeutic interventions targeting ghrelin signaling.
Collapse
Affiliation(s)
| | - Tasha R Davis
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | - James L Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
28
|
Viltart O, Duriez P, Tolle V. Metabolic and neuroendocrine adaptations to undernutrition in anorexia nervosa: from a clinical to a basic research point of view. Horm Mol Biol Clin Investig 2018; 36:hmbci-2018-0010. [PMID: 29804101 DOI: 10.1515/hmbci-2018-0010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
The exact mechanisms linking metabolic and neuroendocrine adaptations to undernutrition and the pathophysiology of anorexia nervosa (AN) are not fully understood. AN is a psychiatric disorder of complex etiology characterized by extreme starvation while the disease is progressing into a chronic state. Metabolic and endocrine alterations associated to this disorder are part of a powerful response to maintain whole body energy homeostasis. But these modifications may also contribute to associated neuropsychiatric symptoms (reward abnormalities, anxiety, depression) and thus participate to sustain the disease. The current review presents data with both a clinical and basic research point of view on the role of nutritional and energy sensors with neuroendocrine actions in the pathophysiology of the disease, as they modulate metabolic responses, reproductive functions, stress responses as well as physical activity. While clinical data present a full description of changes occurring in AN, animal models that integrate either spontaneous genetic mutations or experimentally-induced food restriction with hyperactivity and/or social stress recapitulate the main metabolic and endocrine alterations of AN and provide mechanistic information between undernutrition state and symptoms of the disease. Further progress on the central and peripheral mechanism involved in the pathophysiology of eating disorders partly relies on the development and/or refinement of existing animal models to include recently identified genetic traits and better mimic the complex and multifactorial dimensions of the disease.
Collapse
Affiliation(s)
- Odile Viltart
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Université de Lille (Sciences et technologies), Lille, France
| | - Philibert Duriez
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Clinique des Maladies Mentales et de l'Encéphale (CMME), Hôpital Sainte-Anne, Paris, France
| | - Virginie Tolle
- Centre de Psychiatrie et Neurosciences, INSERM UMR 894, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
29
|
Effects of pregnancy and short-lasting acute feed restriction on total ghrelin concentration and metabolic parameters in dairy cattle. Theriogenology 2018; 106:141-148. [DOI: 10.1016/j.theriogenology.2017.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 10/02/2017] [Accepted: 10/07/2017] [Indexed: 02/01/2023]
|
30
|
Mahbod P, Smith EP, Fitzgerald ME, Morano RL, Packard BA, Ghosal S, Scheimann JR, Perez-Tilve D, Herman JP, Tong J. Desacyl Ghrelin Decreases Anxiety-like Behavior in Male Mice. Endocrinology 2018; 159:388-399. [PMID: 29155981 PMCID: PMC5761608 DOI: 10.1210/en.2017-00540] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 11/10/2017] [Indexed: 11/19/2022]
Abstract
Ghrelin is a 28-amino acid polypeptide that regulates feeding, glucose metabolism, and emotionality (stress, anxiety, and depression). Plasma ghrelin circulates as desacyl ghrelin (DAG) or, in an acylated form, acyl ghrelin (AG), through the actions of ghrelin O-acyltransferase (GOAT), exhibiting low or high affinity, respectively, for the growth hormone secretagogue receptor (GHSR) 1a. We investigated the role of endogenous AG, DAG, and GHSR1a signaling on anxiety and stress responses using ghrelin knockout (Ghr KO), GOAT KO, and Ghsr stop-floxed (Ghsr null) mice. Behavioral and hormonal responses were tested in the elevated plus maze and light/dark (LD) box. Mice lacking both AG and DAG (Ghr KO) increased anxiety-like behaviors across tests, whereas anxiety reactions were attenuated in DAG-treated Ghr KO mice and in mice lacking AG (GOAT KO). Notably, loss of GHSR1a (Ghsr null) did not affect anxiety-like behavior in any test. Administration of AG and DAG to Ghr KO mice with lifelong ghrelin deficiency reduced anxiety-like behavior and decreased phospho-extracellular signal-regulated kinase phosphorylation in the Edinger-Westphal nucleus in wild-type mice, a site normally expressing GHSR1a and involved in stress- and anxiety-related behavior. Collectively, our data demonstrate distinct roles for endogenous AG and DAG in regulation of anxiety responses and suggest that the behavioral impact of ghrelin may be context dependent.
Collapse
Affiliation(s)
- Parinaz Mahbod
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Eric P. Smith
- Department of Medicine, University of Cincinnati,
Cincinnati, Ohio 45267
| | - Maureen E. Fitzgerald
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Rachel L. Morano
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Benjamin A. Packard
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Sriparna Ghosal
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Jessie R. Scheimann
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Diego Perez-Tilve
- Department of Medicine, University of Cincinnati,
Cincinnati, Ohio 45267
| | - James P. Herman
- Department of Psychiatry and Behavioral Neuroscience,
University of Cincinnati, Cincinnati, Ohio 45267
| | - Jenny Tong
- Division of Endocrinology, Metabolism and Nutrition,
Department of Medicine, Duke University, Durham, North Carolina 27708
| |
Collapse
|
31
|
Cleverdon ER, McGovern-Gooch KR, Hougland JL. The octanoylated energy regulating hormone ghrelin: An expanded view of ghrelin's biological interactions and avenues for controlling ghrelin signaling. Mol Membr Biol 2017; 33:111-124. [PMID: 29143554 DOI: 10.1080/09687688.2017.1388930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ghrelin is a small peptide hormone that requires a unique post-translational modification, serine octanoylation, to bind and activate the GHS-R1a receptor. Initially demonstrated to stimulate hunger and appetite, ghrelin-dependent signaling is implicated in a variety of neurological and physiological processes influencing diseases such as diabetes, obesity, and Prader-Willi syndrome. In addition to its cognate receptor, recent studies have revealed ghrelin interacts with a range of binding partners within the bloodstream. Defining the scope of ghrelin's interactions within the body, understanding how these interactions work in concert to modulate ghrelin signaling, and developing molecular tools for controlling ghrelin signaling are essential for exploiting ghrelin for therapeutic effect. In this review, we discuss recent findings regarding the biological effects of ghrelin signaling, outline binding partners that control ghrelin trafficking and stability in circulation, and summarize the current landscape of inhibitors targeting ghrelin octanoylation.
Collapse
Affiliation(s)
| | | | - James L Hougland
- a Department of Chemistry , Syracuse University , Syracuse , NY , USA
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW In recent years, the role of the gastrointestinal (GI) tract in energy homeostasis through modulation of the digestion and absorption of carbohydrates and the production of incretin hormones is well recognized. RECENT FINDINGS Bariatric surgery for obesity has been a very effective method in substantially improving weight, and numerous studies have focused on intestinal adaptation after bariatric procedures. A number of structural and functional changes in the GI tract have been reported postsurgery, which could be responsible for the altered hormonal responses. Furthermore, the change in food absorption rate and the intestinal regions exposed to carbohydrates may affect blood glucose response. This review hopes to give new insights into the direct role of gut hormones, by summarising the metabolic effects of bariatric surgery.
Collapse
Affiliation(s)
- Georgios K Dimitriadis
- Division of Translational and Experimental Medicine, Clinical Sciences Research Laboratories, University of Warwick Medical School, Coventry, CV2 2DX, UK.
- Academic Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, W12 0NN, UK.
- Division of Translational and Experimental Medicine-Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| | - Manpal S Randeva
- Division of Translational and Experimental Medicine, Clinical Sciences Research Laboratories, University of Warwick Medical School, Coventry, CV2 2DX, UK
| | - Alexander D Miras
- Academic Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, W12 0NN, UK
| |
Collapse
|
33
|
Santiago-Fernández C, García-Serrano S, Tome M, Valdes S, Ocaña-Wilhelmi L, Rodríguez-Cañete A, Tinahones FJ, García-Fuentes E, Garrido-Sánchez L. Ghrelin levels could be involved in the improvement of insulin resistance after bariatric surgery. ACTA ACUST UNITED AC 2017; 64:355-362. [PMID: 28745606 DOI: 10.1016/j.endinu.2017.05.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 05/04/2017] [Accepted: 05/11/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVE Ghrelin is a gastrointestinal peptide involved in regulation of body weight and energy balance. However, its behavior after bariatric surgery and its relationship to insulin resistance are still controversial. A simultaneous assessment was made of the association between changes in ghrelin levels and different variables after three types of bariatric surgery. PATIENTS AND METHODS Ghrelin levels were measured in 103 morbidly obese subjects before and 6 months after bariatric surgery (Roux-en-Y gastric bypass (RYGB), biliopancreatic diversion of Scopinaro (BPD), and sleeve gastrectomy (SG)), and in 21 non-obese subjects. RESULTS Ghrelin levels increased after RYGB (p<0.05), were unchanged after BPD, and decreased after SG (p<0.05). The percent change in ghrelin levels (Δ-ghrelin) was associated to the type of surgery in a multiple linear regression model (p=0.017). When the same analysis was only performed in subjects in whom the gastric fundus was maintained (RYGB and BPD), Δ-ghrelin was negatively associated to Δ-HOMA-IR (p=0.001). In morbidly obese subjects who underwent RYGB and BPD, the odds ratio of a lower Δ-HOMA-IR in patients with Δ-ghrelin in the Q1 quartile versus those with Δ-ghrelin in the Q4 quartile was 8.74 (1.73-44.06) (p=0.009). CONCLUSIONS Changes in ghrelin levels after bariatric surgery are associated to the presence or absence of the gastric fundus. After bariatric surgery, the decrease in insulin resistance was associated to increased ghrelin levels in procedures in which the fundus is not excluded.
Collapse
Affiliation(s)
- Concepción Santiago-Fernández
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
| | - Sara García-Serrano
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Malaga, Spain
| | - Mónica Tome
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain
| | - Sergio Valdes
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Malaga, Spain
| | - Luis Ocaña-Wilhelmi
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Transplantes, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Alberto Rodríguez-Cañete
- Unidad de Gestión Clínica de Cirugía General, Digestiva y Transplantes, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario, Málaga, Spain
| | - Francisco J Tinahones
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain.
| | - Eduardo García-Fuentes
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain; Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga, Spain.
| | - Lourdes Garrido-Sánchez
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Málaga, Spain
| |
Collapse
|
34
|
The role of ghrelin-responsive mediobasal hypothalamic neurons in mediating feeding responses to fasting. Mol Metab 2017; 6:882-896. [PMID: 28752052 PMCID: PMC5518774 DOI: 10.1016/j.molmet.2017.06.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 12/20/2022] Open
Abstract
Objective Ghrelin is a stomach-derived hormone that affects food intake and regulates blood glucose. The best-characterized actions of ghrelin are mediated by its binding to and activation of the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Adequate examination of the identity, function, and relevance of specific subsets of GHSR-expressing neurons has been hampered by the absence of a suitable Cre recombinase (Cre)-expressing mouse line with which to manipulate gene expression in a targeted fashion within GHSR-expressing neurons. The present study aims to characterize the functional significance and neurocircuitry of GHSR-expressing neurons in the mediobasal hypothalamus (MBH), as they relate to ghrelin-induced food intake and fasting-associated rebound hyperphagia, using a novel mouse line in which Cre expression is controlled by the Ghsr promoter. Methods A Ghsr-IRES-Cre mouse line that expresses Cre directed by the Ghsr promoter was generated. The line was validated by comparing Cre activity in reporter mice to the known brain distribution pattern of GHSR. Next, the requirement of MBH GHSR-expressing neuronal activity in mediating food intake in response to administered ghrelin and in response to fasting was assessed after stereotaxic delivery of inhibitory designer receptor exclusively activated by designer drugs (DREADD) virus to the MBH. In a separate cohort of Ghsr-IRES-Cre mice, stereotaxic delivery of stimulatory DREADD virus to the MBH was performed to assess the sufficiency of MBH GHSR-expressing neuronal activity on food intake. Finally, the distribution of MBH GHSR-expressing neuronal axonal projections was assessed in the DREADD virus-injected animals. Results The pattern of Cre activity in the Ghsr-IRES-Cre mouse line mostly faithfully reproduced the known GHSR expression pattern. DREADD-assisted inhibition of MBH GHSR neuronal activity robustly suppressed the normal orexigenic response to ghrelin and fasting-associated rebound food intake. DREADD-assisted stimulation of MBH GHSR neuronal activity was sufficient to induce food intake. Axonal projections of GHSR-expressing MBH neurons were observed in a subset of hypothalamic and extra-hypothalamic regions. Conclusions These results suggest that 1) activation of GHSR-expressing neurons in the MBH is required for the normal feeding responses following both peripheral administration of ghrelin and fasting, 2) activation of MBH GHSR-expressing neurons is sufficient to induce feeding, and 3) axonal projections to a subset of hypothalamic and/or extra-hypothalamic regions likely mediate these responses. The Ghsr-IRES-Cre line should serve as a valuable tool to further our understanding of the functional significance of ghrelin-responsive/GHSR-expressing neurons and the neuronal circuitry within which they act. We generated a novel Ghsr-IRES-Cre knock-in mouse line. Cre activity in the line mirrors the known GHSR expression pattern. Chemogenetic modulation of neuronal activity reveals a required role of MBH GHSR neurons in rebound food intake after a fast. Neuronal projections of mediobasal hypothalamic GHSR neurons are reminiscent of AgRP neuronal projections.
Collapse
|
35
|
Cho YH, Lee SY, Jeong DW, Cho AR, Jeon JS, Kim YJ, Lee JG, Yi YH, Tak YJ, Hwang HR, Lee SH, Lee SM. Metabolic Syndrome is Associated with Lower Plasma Levels of Desacyl Ghrelin and Total Ghrelin in Asymptomatic Middle-aged Korean Men. J Obes Metab Syndr 2017; 26:114-121. [PMID: 31089505 PMCID: PMC6484903 DOI: 10.7570/jomes.2017.26.2.114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 10/27/2016] [Accepted: 12/12/2016] [Indexed: 01/08/2023] Open
Abstract
Background Desacyl ghrelin is acylated by ghrelin O-acyltransferase (GOAT) and converted to acyl ghrelin. To date, little is known about the relationship among the levels of these two forms of ghrelin, GOAT level, and insulin resistance in Asian individuals. The purpose of this study was to determine the relationship between insulin resistance and the levels of plasma acyl ghrelin, desacyl ghrelin, and GOAT in asymptomatic middle-aged Korean men. Methods This cross-sectional study evaluated 78 asymptomatic middle-aged Korean men with metabolic syndrome (MS). We examined the correlation between the plasma levels of acyl ghrelin, desacyl ghrelin, and GOAT and sociodemographic, dietary, anthropometric, and metabolic parameters, as well as the association between insulin resistance and plasma levels of acyl ghrelin, desacyl ghrelin, and GOAT. Results The levels of desacyl ghrelin and total ghrelin were significantly lower in the MS group than in the non-MS group (P<0.017, P=0.01, respectively). HOMA-IR values showed a significant negative correlation with desacyl ghrelin (r=−0.271, P=0.017) and total ghrelin (r=−0.271, P=0.016) levels. Acyl ghrelin and GOAT were not significantly correlated with HOMA-IR, and no correlation was found between the plasma levels of the two ghrelin types and GOAT. Conclusion The plasma levels of desacyl ghrelin and total ghrelin in middle-aged Korean men with MS were lower than in those without MS. A significant negative correlation was observed between desacyl ghrelin level and HOMA-IR; however, no correlation was found between plasma levels of acyl ghrelin and GOAT and HOMA-IR.
Collapse
Affiliation(s)
- Young Hye Cho
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sang Yeoup Lee
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea.,Medical Education Unit, Pusan National University, School of Medicine, Yangsan, Korea
| | - Dong Wook Jeong
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - A Ra Cho
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jeong Suk Jeon
- Family Medicine Clinic, Obesity, Metabolism and Nutrition Center and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Yun Jin Kim
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Jeong Gyu Lee
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Yu Hyeon Yi
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Young Jin Tak
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Hye Rim Hwang
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Seung-Hun Lee
- Department of Family Medicine, Pusan National University Hospital, Busan, Korea
| | - Sun Min Lee
- Department of Laboratory Medicine and Molecular Genetics and Research Institute of Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
36
|
Cavin JB, Bado A, Le Gall M. Intestinal Adaptations after Bariatric Surgery: Consequences on Glucose Homeostasis. Trends Endocrinol Metab 2017; 28:354-364. [PMID: 28209316 DOI: 10.1016/j.tem.2017.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/16/2017] [Accepted: 01/17/2017] [Indexed: 12/25/2022]
Abstract
The gastrointestinal (GI) tract can play a direct role in glucose homeostasis by modulating the digestion and absorption of carbohydrates and by producing the incretin hormones. In recent years, numerous studies have focused on intestinal adaptation following bariatric surgeries. Changes in the number of incretin (glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide) producing cells have been reported, which could result in the modified hormonal response seen after surgery. In addition, the rate of absorption and the intestinal regions exposed to sugars may affect the time course of appearance of glucose in the blood. This review gives new insights into the direct role of the GI tract in the metabolic outcomes of bariatric surgery, in the context of glucose homeostasis.
Collapse
Affiliation(s)
- Jean-Baptiste Cavin
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, DHU Unity AP-HP, F-75890 Paris, France
| | - André Bado
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, DHU Unity AP-HP, F-75890 Paris, France
| | - Maude Le Gall
- Inserm UMR 1149, UFR de Médecine Paris Diderot, Université Paris Diderot, Sorbonne Paris Cité, DHU Unity AP-HP, F-75890 Paris, France.
| |
Collapse
|
37
|
Blanco AM, Bertucci JI, Ramesh N, Delgado MJ, Valenciano AI, Unniappan S. Ghrelin Facilitates GLUT2-, SGLT1- and SGLT2-mediated Intestinal Glucose Transport in Goldfish (Carassius auratus). Sci Rep 2017; 7:45024. [PMID: 28338019 PMCID: PMC5364492 DOI: 10.1038/srep45024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/17/2017] [Indexed: 12/13/2022] Open
Abstract
Glucose homeostasis is an important biological process that involves a variety of regulatory mechanisms. This study aimed to determine whether ghrelin, a multifunctional gut-brain hormone, modulates intestinal glucose transport in goldfish (Carassius auratus). Three intestinal glucose transporters, the facilitative glucose transporter 2 (GLUT2), and the sodium/glucose co-transporters 1 (SGLT1) and 2 (SGLT2), were studied. Immunostaining of intestinal sections found colocalization of ghrelin and GLUT2 and SGLT2 in mucosal cells. Some cells containing GLUT2, SGLT1 and SGLT2 coexpressed the ghrelin/growth hormone secretagogue receptor 1a (GHS-R1a). Intraperitoneal glucose administration led to a significant increase in serum ghrelin levels, as well as an upregulation of intestinal preproghrelin, ghrelin O-acyltransferase and ghs-r1 expression. In vivo and in vitro ghrelin treatment caused a concentration- and time-dependent modulation (mainly stimulatory) of GLUT2, SGLT1 and SGLT2. These effects were abolished by the GHS-R1a antagonist [D-Lys3]-GHRP-6 and the phospholipase C inhibitor U73122, suggesting that ghrelin actions on glucose transporters are mediated by GHS-R1a via the PLC/PKC signaling pathway. Finally, ghrelin stimulated the translocation of GLUT2 into the plasma membrane of goldfish primary intestinal cells. Overall, data reported here indicate an important role for ghrelin in the modulation of glucoregulatory machinery and glucose homeostasis in fish.
Collapse
Affiliation(s)
- Ayelén Melisa Blanco
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain.,Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Juan Ignacio Bertucci
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico Chascomús, Buenos Aires, Argentina
| | - Naresh Ramesh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - María Jesús Delgado
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana Isabel Valenciano
- Departamento de Fisiología (Fisiología Animal II), Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
38
|
Labarthe A, Tolle V. [Ghrelin: a gastric hormone at the crossroad between growth and appetite regulation]. Biol Aujourdhui 2017; 210:237-257. [PMID: 28327282 DOI: 10.1051/jbio/2016027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Indexed: 06/06/2023]
Abstract
Ghrelin is a 28 amino acid peptide hormone synthesized within the gastrointestinal tract. Initially identified as the endogenous ligand of the GHS-R1a (Growth Hormone Secretagogue Receptor 1a), ghrelin is a powerful stimulator of growth hormone (GH) secretion. At the crossroad between nutrition, growth and long-term energy metabolism, ghrelin also plays a unique role as the first identified gastric hormone increasing appetite and adiposity. However, the role of the ghrelin/GHS-R system in the physiology of growth, feeding behaviour and energy homeostasis needs to be better understood. Utilization of pharmacological tools and complementary animal models with deficiency in preproghrelin, ghrelin-O-acyl-transferase (GOAT - the enzyme that acylates ghrelin -) or GHS-R in situations of chronic undernutrition or high fat diet gives a more precise overview of the role of ghrelin in the pathophysiology of eating and metabolic disorders.
Collapse
|
39
|
McGovern-Gooch KR, Mahajani NS, Garagozzo A, Schramm AJ, Hannah LG, Sieburg MA, Chisholm JD, Hougland JL. Synthetic Triterpenoid Inhibition of Human Ghrelin O-Acyltransferase: The Involvement of a Functionally Required Cysteine Provides Mechanistic Insight into Ghrelin Acylation. Biochemistry 2017; 56:919-931. [PMID: 28134508 DOI: 10.1021/acs.biochem.6b01008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The peptide hormone ghrelin plays a key role in regulating hunger and energy balance within the body. Ghrelin signaling presents a promising and unexploited target for development of small molecule therapeutics for treatment of obesity, diabetes, and other health conditions. Inhibition of ghrelin O-acyltransferase (GOAT), which catalyzes an essential octanoylation step in ghrelin maturation, offers a potential avenue for controlling ghrelin signaling. Through screening a small molecule library, we have identified a class of synthetic triterpenoids that efficiently inhibit ghrelin acylation by the human isoform of GOAT (hGOAT). These compounds function as covalent reversible inhibitors of hGOAT, providing the first evidence of the involvement of a nucleophilic cysteine residue in substrate acylation by a MBOAT family acyltransferase. Surprisingly, the mouse form of GOAT does not exhibit susceptibility to cysteine-modifying electrophiles, revealing an important distinction in the activity and behavior between these closely related GOAT isoforms. This study establishes these compounds as potent small molecule inhibitors of ghrelin acylation and provides a foundation for the development of novel hGOAT inhibitors as therapeutics targeting diabetes and obesity.
Collapse
Affiliation(s)
| | - Nivedita S Mahajani
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Ariana Garagozzo
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Anthony J Schramm
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Lauren G Hannah
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - Michelle A Sieburg
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - John D Chisholm
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| | - James L Hougland
- Department of Chemistry, Syracuse University , Syracuse, New York 13244, United States
| |
Collapse
|
40
|
Churm R, Davies JS, Stephens JW, Prior SL. Ghrelin function in human obesity and type 2 diabetes: a concise review. Obes Rev 2017; 18:140-148. [PMID: 27899023 DOI: 10.1111/obr.12474] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/12/2022]
Abstract
The 28 amino acid hormone, ghrelin, has been found to have various effects on metabolism. This review will focus on the pathways integrated into ghrelin's effect within the hypothalamus, pancreas and adipocytes. The identification of molecules and pathways that regulate ghrelin-mediated lipid retention could establish new mechanisms underlying cellular energy homeostasis. The impact of acyl-ghrelin on glucose metabolism and lipid homeostasis may allow for novel preventative or early intervention therapeutic strategies to treat obesity related type 2 diabetes and associated metabolic dysfunction.
Collapse
Affiliation(s)
- R Churm
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - J S Davies
- Molecular Neurobiology Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - J W Stephens
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| | - S L Prior
- Diabetes Research Group, Institute of Life Science 1, Swansea University, Swansea, UK
| |
Collapse
|
41
|
Yu TY, Wei JN, Kuo CH, Liou JM, Lin MS, Shih SR, Hua CH, Hsein YC, Hsu YW, Chuang LM, Lee MK, Hsiao CH, Wu MS, Li HY. The Impact of Gastric Atrophy on the Incidence of Diabetes. Sci Rep 2017; 7:39777. [PMID: 28045079 PMCID: PMC5206635 DOI: 10.1038/srep39777] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 11/28/2016] [Indexed: 01/08/2023] Open
Abstract
Gastric atrophy results in lower plasma ghrelin, higher gastrin secretion, a change in gut microbiota, and altered dietary nutrient absorption, which may be associated with the incidence of diabetes. Helicobacter pylori (H. pylori) infection is a major cause of gastric atrophy and is associated with diabetes in some reports. Since there is no study which investigates the impact of gastric atrophy on diabetes, we conduct a prospective cohort study to examine the relationship between H. pylori infection, gastric atrophy, and incident diabetes. In this study, subjects with gastric atrophy had a lower risk of incident diabetes, compared to those without gastric atrophy. The extent of gastric atrophy, measured by serum pepsinogen (PG) I/II ratio, was correlated with age, H. pylori IgG titer, HOMA2-IR, and HOMA2%B. When gastric atrophy is more extensive, presented as a lower serum PG I/II ratio, the risk of incident diabetes is lower. On the other hand, there was no significant association between H. pylori infection and the incidence of diabetes. In conclusion, the presence and the extent of gastric atrophy, but not H. pylori infection, are associated with incident diabetes. Further studies are needed to investigate the detailed mechanisms and the potential applications of the findings to guide diabetes screening and treatment strategies.
Collapse
Affiliation(s)
- Tse-Ya Yu
- Health Management Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jung-Nan Wei
- Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chun-Heng Kuo
- Department of Internal Medicine, New Taipei City Hospital, New Taipei City, Taiwan
| | - Jyh-Ming Liou
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mao-Shin Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Shyang-Rong Shih
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Cyue-Huei Hua
- Division of Clinical Pathology' National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Yenh-Chen Hsein
- Division of Clinical Pathology' National Taiwan University Hospital Yun-Lin Branch, Yun-Lin, Taiwan
| | - Ya-Wen Hsu
- Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Kuei Lee
- Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | | | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Yuan Li
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
42
|
Pereira JADS, da Silva FC, de Moraes-Vieira PMM. The Impact of Ghrelin in Metabolic Diseases: An Immune Perspective. J Diabetes Res 2017; 2017:4527980. [PMID: 29082258 PMCID: PMC5610818 DOI: 10.1155/2017/4527980] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 07/07/2017] [Accepted: 07/31/2017] [Indexed: 01/22/2023] Open
Abstract
Obesity and insulin resistance have reached epidemic proportions. Obesogenic conditions are associated with increased risk for the development of other comorbidities and obesity-related diseases. In metabolic disorders, there is chronic low-grade inflammation induced by the activation of immune cells, especially in metabolic relevant organs such as white adipose tissue (WAT). These immune cells are regulated by environmental and systemic cues. Ghrelin is a peptide secreted mainly by X/A-like gastric cells and acts through the growth hormone secretagogue receptor (GHS-R). This receptor is broadly expressed in the central nervous system (CNS) and in several cell types, including immune cells. Studies show that ghrelin induces an orexigenic state, and there is increasing evidence implicating an immunoregulatory role for ghrelin. Ghrelin mainly acts on the innate and adaptive immune systems to suppress inflammation and induce an anti-inflammatory profile. In this review, we discuss the immunoregulatory roles of ghrelin, the mechanisms by which ghrelin acts and potential pharmacological applications for ghrelin in the treatment of obesity-associated inflammatory diseases, such as type 2 diabetes (T2D).
Collapse
Affiliation(s)
- Jéssica Aparecida da Silva Pereira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| | - Felipe Corrêa da Silva
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
| | - Pedro Manoel Mendes de Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution and Bioagents, Institute of Biology, University of Campinas, São Paulo, SP, Brazil
- Department of Immunology, Institute of Biomedical Science, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
43
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 392] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
44
|
Villa-Osaba A, Gahete MD, Cordoba-Chacon J, de Lecea L, Castaño JP, Luque RM. Fasting modulates GH/IGF-I axis and its regulatory systems in the mammary gland of female mice: Influence of endogenous cortistatin. Mol Cell Endocrinol 2016; 434:14-24. [PMID: 27291340 DOI: 10.1016/j.mce.2016.06.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/27/2016] [Accepted: 06/08/2016] [Indexed: 11/19/2022]
Abstract
Growth hormone (GH) and insulin-like growth factor-I (IGF-I) are essential factors in mammary-gland (MG) development and are altered during fasting. However, no studies have investigated the alterations in the expression of GH/IGF-I and its regulatory systems (somatostatin/cortistatin and ghrelin) in MG during fasting. Therefore, this study was aimed at characterizing the regulation of GH/IGF-I/somatostatin/cortistatin/ghrelin-systems expression in MG of fasted female-mice (compared to fed-controls) and the influence of endogenous-cortistatin (using cortistatin-knockouts). Fasting decreased IGF-I while increased IGF-I/Insulin-receptors expression in MGs. Fasting provoked an increase in GH expression that might be associated to enhanced ghrelin-variants/ghrelin-O-acyl-transferase enzyme expression, while an upregulation of somatostatin-receptors was observed. However, cortistatin-knockouts mice showed a decrease in GH and somatostatin receptor-subtypes expression. Altogether, we demonstrate that GH/IGF-I, somatostatin/cortistatin and ghrelin systems expression is altered in MG during fasting, suggesting a relevant role in coordinating its response to metabolic stress, wherein endogenous cortistatin might be essential for an appropriate response.
Collapse
Affiliation(s)
- Alicia Villa-Osaba
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - José Cordoba-Chacon
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain.
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Hospital Universitario Reina Sofía (HURS), Spain; CIBERobn, Córdoba, Spain; ceiA3, Córdoba, Spain.
| |
Collapse
|
45
|
Luvuno M, Mbongwa HP, Khathi A. THE EFFECTS OF Syzygium aromaticum-DERIVED TRITERPENES ON GASTROINTESTINAL GHRELIN EXPRESSION IN STREPTOZOTOCIN-INDUCED DIABETIC RATS. AFRICAN JOURNAL OF TRADITIONAL, COMPLEMENTARY, AND ALTERNATIVE MEDICINES 2016; 13:8-14. [PMID: 28852714 PMCID: PMC5566155 DOI: 10.21010/ajtcam.v13i4.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Diabetic polyphagia has been associated with elevated plasma ghrelin levels in experimental type 1 diabetes. This increase in food consumption contributes to chronic hyperglycaemia in diabetes thus contributing to the development of micro- and macrovascular complications. We have reported that plant-derived oleanolic acid (OA) and maslinic acid (MA) reduce blood glucose levels, in part, through the inhibition of intestinal carbohydrate hydrolyzing enzymes and glucose transporters. However, their effects on food intake and plasma ghrelin concentrations are unclear. Accordingly, we investigated the effects of these triterpenes on food intake and ghrelin expression in streptozotocin-induced diabetic rats. Material: The effects of OA and MA on blood glucose concentration; food and water intake were monitored over five weeks after which plasma ghrelin concentrations were measured. Additionally, the expression of ghrelin in the various sections of the GIT was determined using Western blot analysis. Results: Ghrelin concentrations in untreated STZ-induced diabetic rats were significantly higher in comparison to the non-diabetic control. Interestingly, the administration of OA and MA reduced food intake, blood glucose levels and plasma ghrelin levels in STZ-induced diabetic rats. This was further complemented by significant reductions in the gastrointestinal expression of ghrelin suggesting that the anti-diabetic properties of these triterpenes are mediated, in part, through the reduction of food intake and the modulation of ghrelin expression. Conclusion: The findings of the study suggest that the control of food intake through the reduction of ghrelin expression by plant-derived OA and MA may constitute an avenue of glycaemic control in diabetes mellitus.
Collapse
Affiliation(s)
- Mluleki Luvuno
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Hlengiwe Prosperity Mbongwa
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Andile Khathi
- Department of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
46
|
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: 0.3] [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
|
47
|
Affiliation(s)
| | - William Snape
- California Pacific Medical Center, San Francisco, California
| |
Collapse
|
48
|
Naftalin RJ. A computer model simulating human glucose absorption and metabolism in health and metabolic disease states. F1000Res 2016; 5:647. [PMID: 27347379 PMCID: PMC4909112 DOI: 10.12688/f1000research.8299.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
A computer model designed to simulate integrated glucose-dependent changes in splanchnic blood flow with small intestinal glucose absorption, hormonal and incretin circulation and hepatic and systemic metabolism in health and metabolic diseases e.g. non-alcoholic fatty liver disease, (NAFLD), non-alcoholic steatohepatitis, (NASH) and type 2 diabetes mellitus, (T2DM) demonstrates how when glucagon-like peptide-1, (GLP-1) is synchronously released into the splanchnic blood during intestinal glucose absorption, it stimulates superior mesenteric arterial (SMA) blood flow and by increasing passive intestinal glucose absorption, harmonizes absorption with its distribution and metabolism. GLP-1 also synergises insulin-dependent net hepatic glucose uptake (NHGU). When GLP-1 secretion is deficient post-prandial SMA blood flow is not increased and as NHGU is also reduced, hyperglycaemia follows. Portal venous glucose concentration is also raised, thereby retarding the passive component of intestinal glucose absorption. Increased pre-hepatic sinusoidal resistance combined with portal hypertension leading to opening of intrahepatic portosystemic collateral vessels are NASH-related mechanical defects that alter the balance between splanchnic and systemic distributions of glucose, hormones and incretins.The model reveals the latent contribution of portosystemic shunting in development of metabolic disease. This diverts splanchnic blood content away from the hepatic sinuses to the systemic circulation, particularly during the glucose absorptive phase of digestion, resulting in inappropriate increases in insulin-dependent systemic glucose metabolism. This hastens onset of hypoglycaemia and thence hyperglucagonaemia. The model reveals that low rates of GLP-1 secretion, frequently associated with T2DM and NASH, may be also be caused by splanchnic hypoglycaemia, rather than to intrinsic loss of incretin secretory capacity. These findings may have therapeutic implications on GLP-1 agonist or glucagon antagonist usage.
Collapse
Affiliation(s)
- Richard J Naftalin
- Departments of Physiology and Vascular Biology, BHF centre of research excellence, King's College London School of Medicine, London, UK
| |
Collapse
|
49
|
Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides 2016; 77:28-37. [PMID: 26344355 DOI: 10.1016/j.peptides.2015.08.013] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 12/17/2022]
Abstract
Bariatric surgery for obesity has proved to be an extremely effective method of promoting long-term weight reduction with additional beneficial metabolic effects, such as improved glucose tolerance and remission of type 2 diabetes. A range of bariatric procedures are in common use, including gastric banding, sleeve gastrectomy and the Roux-en-Y gastric bypass. Although the mechanisms underlying the efficacy of bariatric surgery are unclear, gastrointestinal and pancreatic peptides are thought to play an important role. The aim of this review is to summarise the effects of different bariatric surgery procedures upon gastrointestinal and pancreatic peptides, including ghrelin, gastrin, cholecystokinin (CCK), glucose-dependent insulinotropic hormone (GIP), glucagon-like peptide 1 (GLP-1), peptide YY (PYY), oxyntomodulin, insulin, glucagon and somatostatin.
Collapse
Affiliation(s)
- Claire L Meek
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Hannah B Lewis
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Frank Reimann
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Fiona M Gribble
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Adrian J Park
- Institute of Metabolic Science, Metabolic Research Laboratories, University of Cambridge, Addenbrookes's Hospital, Box 289, Hills Road, Cambridge CB2 0QQ, United Kingdom; Department of Clinical Biochemistry, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
50
|
Sharma R, Hassan C, Chaiban JT. Severe Insulin Resistance Improves Immediately After Sleeve Gastrectomy. J Investig Med High Impact Case Rep 2016; 4:2324709615625309. [PMID: 26788532 PMCID: PMC4710130 DOI: 10.1177/2324709615625309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 12/08/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction. Obese individuals exhibit insulin resistance often leading to adverse health outcomes. When compared with intensive medical therapy, bariatric surgery has shown better outcomes mainly in terms of insulin resistance and glycemic control. Using the Homeostasis Model Assessment of insulin resistance (HOMA-IR), we report herein a case illustrating a drastic improvement in severe insulin resistance after sleeve gastrectomy in the immediate postoperative period. Case Report. A patient with long-standing history of morbid obesity, type 2 diabetes, obstructive sleep apnea, hypertension, and severe insulin resistance (requiring approximately 2 units of insulin per kg per day) was enrolled in the medical weight management program for 6 months during which he lost 40 lbs and his insulin requirements decreased. He then underwent a sleeve gastrectomy and did not require insulin therapy as of postoperative day 1. His HOMA-IR improved by about 76% between day 1 and day 14 postoperatively. Conclusion. Sleeve gastrectomy leads to a drastic improvement in severe insulin resistance as early as the first postoperative day.
Collapse
Affiliation(s)
- Rahul Sharma
- Case Western Reserve University, Cleveland, OH, USA; St Vincent Charity Medical Center, Cleveland, OH, USA
| | | | - Joumana T Chaiban
- Case Western Reserve University, Cleveland, OH, USA; St Vincent Charity Medical Center, Cleveland, OH, USA
| |
Collapse
|