1
|
Hua D, Huang W, Xu W, Zhang Y, Xie Q, Li P, Sheng Y. Targeting of G protein-coupled receptor 39 alleviates angiotensin II-induced renal damage by reducing ribonucleotide reductase M2. Exp Cell Res 2024; 440:114102. [PMID: 38821252 DOI: 10.1016/j.yexcr.2024.114102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024]
Abstract
Renal fibrosis, apoptosis and autophagy are the main pathological manifestations of angiotensin II (Ang II)-induced renal injury. G protein-coupled receptor 39 (GPR39) is highly expressed in various tissues including the kidney, but its role in the kidney is entirely unclear. This study was performed to investigate the underlying mechanism by which knockdown of GPR39 alleviated Ang II-induced renal injury. In vivo, GPR39 knockout (KO) mice were constructed and infused with Ang II for 4 weeks, followed by renal function tests. In vitro, Ang II-induced cells were treated with si-GPR39 for 48 h. Fibrosis, apoptosis and autophagy were detected in both cells and mice. The underlying mechanism was sought by mRNA transcriptome sequencing and validated in vitro. GPR39 was upregulated in renal tissues of mice with Ang II-mediated renal injury. Knockdown of GPR39 ameliorated renal fibrosis, apoptosis, and autophagy, and decreased the expression of ribonucleotide reductase M2 (RRM2). In vitro, knockdown of GPR39 was also identified to improve the Ang II-induced cell fibrosis, apoptosis, and autophagy. mRNA transcriptome results showed that knockout of GPR39 reduced the expression of RRM2 in Ang II-induced kidney tissue. Activation of RRM2 could reverse the therapeutic effect of GPR39 knockout, and the inhibitor of RRM2 could improve the cell fibrosis, apoptosis and autophagy caused by GPR39 agonist. These results indicated that targeting of GPR39 could alleviate Ang II-induced renal fibrosis, apoptosis, and autophagy via reduction of RRM2 expression, and GPR39 may serve as a potential target for Ang II-induced renal injury.
Collapse
Affiliation(s)
- Dongxu Hua
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, PR China; Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Wanlin Huang
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, PR China
| | - Wenna Xu
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, PR China
| | - Yue Zhang
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China; Department of Cardiology, Jiangsu Province People's Hospital, Nanjing, Jiangsu, PR China
| | - Qiyang Xie
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China; Department of Cardiology, Jiangsu Province People's Hospital, Nanjing, Jiangsu, PR China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, PR China; Department of Cardiology, Jiangsu Province People's Hospital, Nanjing, Jiangsu, PR China; Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, PR China.
| | - Yanhui Sheng
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, PR China; Department of Cardiology, Jiangsu Province People's Hospital, Nanjing, Jiangsu, PR China.
| |
Collapse
|
2
|
Zi Z, Rao Y. Discoveries of GPR39 as an evolutionarily conserved receptor for bile acids and of its involvement in biliary acute pancreatitis. SCIENCE ADVANCES 2024; 10:eadj0146. [PMID: 38306436 PMCID: PMC10836733 DOI: 10.1126/sciadv.adj0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/04/2024] [Indexed: 02/04/2024]
Abstract
Acute pancreatitis (AP) is one of the most common gastrointestinal diseases. Bile acids (BAs) were proposed to be a cause of AP nearly 170 years ago, though the underlying mechanisms remain unclear. Here, we report that two G protein-coupled receptors, GPR39 and GHSR, mediated cellular responses to BAs. Our results revealed GPR39 as an evolutionarily conserved receptor for BAs, particularly 3-O-sulfated lithocholic acids. In cultured cell lines, GPR39 is sufficient for BA-induced Ca2+ elevation. In pancreatic acinar cells, GPR39 mediated BA-induced Ca2+ elevation and necrosis. Furthermore, AP induced by BAs was significantly reduced in GPR39 knockout mice. Our findings provide in vitro and in vivo evidence demonstrating that GPR39 is necessary and sufficient to mediate BA signaling, highlighting its involvement in biliary AP pathogenesis, and suggesting it as a promising therapeutic target for biliary AP.
Collapse
Affiliation(s)
- Zhentao Zi
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yi Rao
- Chinese Institutes for Medical Research, Beijing (CIMR, Beijing) and the State Key Laboratory of Digestive Health, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, PKU-IDG/McGovern Institute for Brain Research, School of Life Sciences, School of Pharmaceutical Sciences, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Changping Laboratory, Chinese Institute of Brain Research Beijing and Research Unit of Medical Neurobiology, Chinese Academy of Medical Sciences, Beijing 102206, China
| |
Collapse
|
3
|
Bora RR, Prasad R, Khatib MN. Cardio-Protective Role of a Gut Hormone Obestatin: A Narrative Review. Cureus 2023; 15:e37972. [PMID: 37223200 PMCID: PMC10202687 DOI: 10.7759/cureus.37972] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 04/22/2023] [Indexed: 05/25/2023] Open
Abstract
Obestatin is a gut hormone composed of 23 amino acids that play a role in protecting the heart. It is synthesized from the same preproghrelin gut hormone gene as another gut hormone. The function and receptor of obestatin remain controversial, despite being present in various organs such as the liver, heart, mammary gland, pancreas, and more. The activity of obestatin is opposite to that of ghrelin, another hormone. The GPR-39 receptor is used by obestatin to exert its effects. Obestatin's cardioprotective role can be attributed to its ability to affect various factors, including adipose tissue, blood pressure regulation, heart, ischemia-reperfusion injury, endothelial cells, and diabetes. Because these factors are related to the cardiovascular system, modifying them via obestatin can provide cardioprotection. Furthermore, ghrelin, its antagonist hormone, regulates cardiovascular health. Diabetes mellitus, hypertension, and ischemia-reperfusion injury can all alter ghrelin/obestatin levels. Obestatin has also been shown to impact other organs, reducing weight and appetite, inhibiting food intake, and increasing adipogenesis. Obestatin has a brief half-life and is quickly degraded by proteases in the blood, liver, and kidneys after entering circulation. This article offers insights into the cardiac function of obestatin.
Collapse
Affiliation(s)
- Rajal R Bora
- Physiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Roshan Prasad
- Medicine and Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Mahalaqua Nazli Khatib
- Epidemiology and Public Health, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
4
|
Functions of the Zinc-Sensing Receptor GPR39 in Regulating Intestinal Health in Animals. Int J Mol Sci 2022; 23:ijms232012133. [PMID: 36292986 PMCID: PMC9602648 DOI: 10.3390/ijms232012133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/08/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
G protein-coupled receptor 39 (GPR39) is a zinc-sensing receptor (ZnR) that can sense changes in extracellular Zn2+, mediate Zn2+ signal transmission, and participate in the regulation of numerous physiological activities in living organisms. For example, GPR39 activates the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) and phosphatidylinositol3-kinase/protein kinase B (PI3K/AKT) signaling pathways upon Zn2+ stimulation, enhances the proliferation and differentiation of colonic cells, and regulates ion transport, as well as exerting other functions. In recent years, with the increased attention to animal gut health issues and the intensive research on GPR39, GPR39 has become a potential target for regulating animal intestinal health. On the one hand, GPR39 is involved in regulating ion transport in the animal intestine, mediating the Cl− efflux by activating the K+/Cl− synergistic protein transporter, and relieving diarrhea symptoms. On the other hand, GPR39 can maintain the homeostasis of the animal intestine, promoting pH restoration in colonic cells, regulating gastric acid secretion, and facilitating nutrient absorption. In addition, GPR39 can affect the expression of tight junction proteins in intestinal epithelial cells, improving the barrier function of the animal intestinal mucosa, and maintaining the integrity of the intestine. This review summarizes the structure and signaling transduction processes involving GPR39 and the effect of GPR39 on the regulation of intestinal health in animals, with the aim of further highlighting the role of GPR39 in regulating animal intestinal health and providing new directions and ideas for studying the prevention and treatment of animal intestinal diseases.
Collapse
|
5
|
Molecular Mechanisms and Health Benefits of Ghrelin: A Narrative Review. Nutrients 2022; 14:nu14194191. [PMID: 36235843 PMCID: PMC9572668 DOI: 10.3390/nu14194191] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/26/2022] [Accepted: 10/05/2022] [Indexed: 11/24/2022] Open
Abstract
Ghrelin, an endogenous brain-gut peptide, is secreted in large quantities, mainly from the stomach, in humans and rodents. It can perform the biological function of activating the growth hormone secretagogue receptor (GHSR). Since its discovery in 1999, ample research has focused on promoting its effects on the human appetite and pleasure-reward eating. Extensive, in-depth studies have shown that ghrelin is widely secreted and distributed in tissues. Its role in neurohumoral regulation, such as metabolic homeostasis, inflammation, cardiovascular regulation, anxiety and depression, and advanced cancer cachexia, has attracted increasing attention. However, the effects and regulatory mechanisms of ghrelin on obesity, gastrointestinal (GI) inflammation, cardiovascular disease, stress regulation, cachexia treatment, and the prognosis of advanced cancer have not been fully summarized. This review summarizes ghrelin's numerous effects in participating in a variety of biochemical pathways and the clinical significance of ghrelin in the regulation of the homeostasis of organisms. In addition, potential mechanisms are also introduced.
Collapse
|
6
|
Bah TM, Allen EM, Garcia-Jaramillo M, Perez R, Zarnegarnia Y, Davis CM, Bloom MB, Magana AA, Choi J, Bobe G, Pike MM, Raber J, Maier CS, Alkayed NJ. GPR39 Deficiency Impairs Memory and Alters Oxylipins and Inflammatory Cytokines Without Affecting Cerebral Blood Flow in a High-Fat Diet Mouse Model of Cognitive Impairment. Front Cell Neurosci 2022; 16:893030. [PMID: 35875352 PMCID: PMC9298837 DOI: 10.3389/fncel.2022.893030] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 12/30/2022] Open
Abstract
Vascular cognitive impairment (VCI) is the second most common cause of dementia. There is no treatment for VCI, in part due to a lack of understanding of the underlying mechanisms. The G-protein coupled receptor 39 (GPR39) is regulated by arachidonic acid (AA)-derived oxylipins that have been implicated in VCI. Furthermore, GPR39 is increased in microglia of post mortem human brains with VCI. Carriers of homozygous GPR39 SNPs have a higher burden of white matter hyperintensity, an MRI marker of VCI. We tested the hypothesis that GPR39 plays a protective role against high-fat diet (HFD)-induced cognitive impairment, in part mediated via oxylipins actions on cerebral blood flow (CBF) and neuroinflammation. Homozygous (KO) and heterozygous (Het) GPR39 knockout mice and wild-type (WT) littermates with and without HFD for 8 months were tested for cognitive performance using the novel object recognition (NOR) and the Morris water maze (MWM) tests, followed by CBF measurements using MRI. Brain tissue and plasma oxylipins were quantified with high-performance liquid chromatography coupled to mass spectrometry. Cytokines and chemokines were measured using a multiplex assay. KO mice, regardless of diet, swam further away from platform location in the MWM compared to WT and Het mice. In the NOR test, there were no effects of genotype or diet. Brain and plasma AA-derived oxylipins formed by 11- and 15-lipoxygenase (LOX), cyclooxygenase (COX) and non-enzymatically were increased by HFD and GPR39 deletion. Interleukin-10 (IL-10) was lower in KO mice on HFD than standard diet (STD), whereas IL-4, interferon γ-induced protein-10 (IP-10) and monocyte chemotactic protein-3 (MCP-3) were altered by diet in both WT and KO, but were not affected by genotype. Resting CBF was reduced in WT and KO mice on HFD, with no change in vasoreactivity. The deletion of GPR39 did not change CBF compared to WT mice on either STD or HFD. We conclude that GPR39 plays a role in spatial memory retention and protects against HFD-induced cognitive impairment in part by modulating inflammation and AA-derived oxylipins. The results indicate that GPR39 and oxylipin pathways play a role and may serve as therapeutic targets in VCI.
Collapse
Affiliation(s)
- Thierno M. Bah
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Elyse M. Allen
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Manuel Garcia-Jaramillo
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Yalda Zarnegarnia
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Catherine M. Davis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| | - Madeline B. Bloom
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Armando A. Magana
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Martin M. Pike
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology, Radiation Medicine, and Psychiatry, Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Portland, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Nabil J. Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
7
|
Doboszewska U, Sawicki J, Sajnóg A, Szopa A, Serefko A, Socała K, Pieróg M, Nieoczym D, Mlyniec K, Nowak G, Barałkiewicz D, Sowa I, Wlaź P. Alterations of Serum Magnesium Concentration in Animal Models of Seizures and Epilepsy—The Effects of Treatment with a GPR39 Agonist and Knockout of the Gpr39 Gene. Cells 2022; 11:cells11131987. [PMID: 35805072 PMCID: PMC9265460 DOI: 10.3390/cells11131987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 12/10/2022] Open
Abstract
Several ligands have been proposed for the GPR39 receptor, including the element zinc. The relationship between GPR39 and magnesium homeostasis has not yet been examined, nor has such a relationship in the context of seizures/epilepsy. We used samples from mice that were treated with an agonist of the GPR39 receptor (TC-G 1008) and underwent acute seizures (maximal electroshock (MES)- or 6-hertz-induced seizures) or a chronic, pentylenetetrazole (PTZ)-induced kindling model of epilepsy. MES seizures and PTZ kindling, unlike 6 Hz seizures, increased serum magnesium concentration. In turn, Gpr39-KO mice that underwent PTZ kindling displayed decreased concentrations of this element in serum, compared to WT mice subjected to this procedure. However, the levels of expression of TRPM7 and SlC41A1 proteins—which are responsible for magnesium transport into and out of cells, respectively—did not differ in the hippocampus between Gpr39-KO and WT mice. Furthermore, laser ablation inductively coupled plasma mass spectrometry applied to hippocampal slices did not reveal differences in magnesium levels between the groups. These data show the relationship between magnesium homeostasis and certain types of acute or chronic seizures (MES seizures or PTZ kindling, respectively), but do not explicitly support the role of GPR39 in mediating magnesium balance in the hippocampus in the latter model. However, decreased expression of TRPM7 and increased expression of SLC41A1—which were observed in the hippocampi of Gpr39-KO mice treated with TC-G 1008, in comparison to WT mice that received the same treatment—implicitly support the link between GPR39 and hippocampal magnesium homeostasis.
Collapse
Affiliation(s)
- Urszula Doboszewska
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland; (K.S.); (M.P.); (D.N.); (P.W.)
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland; (K.M.); (G.N.)
- Correspondence: or ; Tel.: +48-81-537-50-10; Fax: +48-81-537-59-01
| | - Jan Sawicki
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, PL 20-093 Lublin, Poland; (J.S.); (I.S.)
| | - Adam Sajnóg
- Department of Trace Analysis, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, PL 61-614 Poznan, Poland; (A.S.); (D.B.)
| | - Aleksandra Szopa
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland; (A.S.); (A.S.)
| | - Anna Serefko
- Chair and Department of Applied and Social Pharmacy, Laboratory of Preclinical Testing, Medical University of Lublin, Chodźki 1, PL 20-093 Lublin, Poland; (A.S.); (A.S.)
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland; (K.S.); (M.P.); (D.N.); (P.W.)
| | - Mateusz Pieróg
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland; (K.S.); (M.P.); (D.N.); (P.W.)
| | - Dorota Nieoczym
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland; (K.S.); (M.P.); (D.N.); (P.W.)
| | - Katarzyna Mlyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland; (K.M.); (G.N.)
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL 30-688 Kraków, Poland; (K.M.); (G.N.)
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smetna 12, PL 31-343 Kraków, Poland
| | - Danuta Barałkiewicz
- Department of Trace Analysis, Adam Mickiewicz University, Uniwersytetu Poznanskiego 8, PL 61-614 Poznan, Poland; (A.S.); (D.B.)
| | - Ireneusz Sowa
- Department of Analytical Chemistry, Medical University of Lublin, Chodźki 4A, PL 20-093 Lublin, Poland; (J.S.); (I.S.)
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033 Lublin, Poland; (K.S.); (M.P.); (D.N.); (P.W.)
| |
Collapse
|
8
|
Alkayed NJ, Cao Z, Qian ZY, Nagarajan S, Liu X, Nelson JW, Xie F, Li B, Fan W, Liu L, Grafe MR, Davis CM, Xiao X, Barnes AP, Kaul S. Control of Coronary Vascular Resistance by Eicosanoids via a Novel GPCR. Am J Physiol Cell Physiol 2022; 322:C1011-C1021. [PMID: 35385329 PMCID: PMC9255704 DOI: 10.1152/ajpcell.00454.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Arachidonic acid metabolites epoxyeicosatrienoates (EETs) and hydroxyeicosatetraenoates (HETEs) are important regulators of myocardial blood flow and coronary vascular resistance (CVR), but their mechanisms of action are not fully understood. We applied a chemoproteomics strategy using a clickable photoaffinity probe to identify G protein coupled receptor 39 (GPR39) as a microvascular smooth muscle cell (mVSMC) receptor selective for two endogenous eicosanoids, 15-HETE and 14,15-EET, which act on the receptor to oppose each other's activity. The former increases mVSMC intracellular calcium via GPR39 and augments coronary microvascular resistance, and the latter inhibits these actions. Furthermore, we find that the efficacy of both ligands is potentiated by zinc acting as an allosteric modulator. Measurements of coronary perfusion pressure (CPP) in GPR39-null hearts using the Langendorff preparation indicate the receptor senses these eicosanoids to regulate microvascular tone. These results implicate GPR39 as an eicosanoid receptor and key regulator of myocardial tissue perfusion. Our findings will have a major impact on understanding the roles of eicosanoids in cardiovascular physiology and disease and provide an opportunity for the development of novel GPR39-targeting therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Nabil J Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Zhiping Cao
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Zu Yuan Qian
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Shanthi Nagarajan
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States.,Medicinal Chemistry Core, Oregon Health & Science University, Portland, Oregon, United States
| | - Xuehong Liu
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Jonathan W Nelson
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Fuchun Xie
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon, United States
| | - Bingbing Li
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon, United States
| | - Wei Fan
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Lijuan Liu
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Marjorie R Grafe
- DDepartment of Pathology, Oregon Health & Science University, Portland, Oregon, United States
| | - Catherine M Davis
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Xiangshu Xiao
- Department of Physiology and Pharmacology, Oregon Health & Science University, Portland, Oregon, United States.,The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Anthony P Barnes
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| | - Sanjiv Kaul
- The Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States
| |
Collapse
|
9
|
Le Duc D, Velluva A, Cassatt-Johnstone M, Olsen RA, Baleka S, Lin CC, Lemke JR, Southon JR, Burdin A, Wang MS, Grunewald S, Rosendahl W, Joger U, Rutschmann S, Hildebrandt TB, Fritsch G, Estes JA, Kelso J, Dalén L, Hofreiter M, Shapiro B, Schöneberg T. Genomic basis for skin phenotype and cold adaptation in the extinct Steller's sea cow. SCIENCE ADVANCES 2022; 8:eabl6496. [PMID: 35119923 PMCID: PMC8816345 DOI: 10.1126/sciadv.abl6496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Steller's sea cow, an extinct sirenian and one of the largest Quaternary mammals, was described by Georg Steller in 1741 and eradicated by humans within 27 years. Here, we complement Steller's descriptions with paleogenomic data from 12 individuals. We identified convergent evolution between Steller's sea cow and cetaceans but not extant sirenians, suggesting a role of several genes in adaptation to cold aquatic (or marine) environments. Among these are inactivations of lipoxygenase genes, which in humans and mouse models cause ichthyosis, a skin disease characterized by a thick, hyperkeratotic epidermis that recapitulates Steller's sea cows' reportedly bark-like skin. We also found that Steller's sea cows' abundance was continuously declining for tens of thousands of years before their description, implying that environmental changes also contributed to their extinction.
Collapse
Affiliation(s)
- Diana Le Duc
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Akhil Velluva
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| | - Molly Cassatt-Johnstone
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Remi-Andre Olsen
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Box 1031 , SE-17121 Solna, Sweden
| | - Sina Baleka
- Evolutionary Adaptive Genomics, Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
- Faculty of Life and Environmental Sciences, University of Iceland, 102 Reykjavik, Iceland
| | - Chen-Ching Lin
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, 11221 Taipei, Taiwan
| | - Johannes R. Lemke
- Institute of Human Genetics, University Medical Center Leipzig, 04103 Leipzig, Germany
| | - John R. Southon
- Keck-CCAMS Group, Earth System Science Department, University of California, Irvine, Irvine, CA 92697, USA
| | - Alexander Burdin
- Kamchatka Branch of Pacific Geographical Institute, Russian Academy of Science, 683000 Petropavlovsk-Kamchatsky, Russia
| | - Ming-Shan Wang
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Howard Hughes Medical Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sonja Grunewald
- Department of Dermatology, Venerology and Allergology, University Medical Center Leipzig, 04103 Leipzig, Germany
| | - Wilfried Rosendahl
- Reiss-Engelhorn-Museum and Curt-Engelhorn-Centre of Archaeometry, 68159 Mannheim, Germany
| | - Ulrich Joger
- State Museum of Natural History, 38106 Braunschweig, Germany
| | - Sereina Rutschmann
- Evolutionary Adaptive Genomics, Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Thomas B. Hildebrandt
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, 10315 Berlin, Germany
- Faculty of Veterinary Medicine, Free University Berlin, 14195 Berlin, Germany
| | - Guido Fritsch
- Department of Reproduction Management, Leibniz Institute for Zoo and Wildlife Research, 10315 Berlin, Germany
| | - James A. Estes
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Janet Kelso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
| | - Love Dalén
- Centre for Palaeogenetics, SE-106 91 Stockholm, Sweden
- Department of Bioinformatics and Genetics, Swedish Museum of Natural History, SE-104 05 Stockholm, Sweden
- Department of Zoology, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Michael Hofreiter
- Evolutionary Adaptive Genomics, Institute for Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Beth Shapiro
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
- Howard Hughes Medical Institute, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
10
|
Obayemi MJ, Akintayo CO, Oniyide AA, Aturamu A, Badejogbin OC, Atuma CL, Saidi AO, Mahmud H, Olaniyi KS. Protective role of melatonin against adipose-hepatic metabolic comorbidities in experimentally induced obese rat model. PLoS One 2021; 16:e0260546. [PMID: 34879109 PMCID: PMC8654266 DOI: 10.1371/journal.pone.0260546] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Background Adipose and hepatic metabolic dysfunctions are critical comorbidities that
also aggravate insulin resistance in obese individuals. Melatonin is a
low-cost agent and previous studies suggest that its use may promote
metabolic health. However, its effects on some comorbidities associated with
obesity are unknown. Herein, we investigated the hypothesis that melatonin
supplementation would attenuate adipose-hepatic metabolic dysfunction in
high fat diet (HFD)-induced obesity in male Wistar rats. Materials and methods Twenty-four adult male Wistar rats (n = 6/group) were used: Control group
received vehicle (normal saline), obese group received 40% high fat diet,
melatonin-treated group received 4 mg/kg of melatonin, and obese plus
melatonin group received 40% HFD and melatonin. The treatment lasted for 12
weeks. Results HFD caused increased food intake, body weight, insulin level, insulin
resistance and plasma and liver lipid but decreased adipose lipid. In
addition, HFD also increased plasma, adipose and liver malondialdehyde,
IL-6, uric acid and decreased Glucose-6-phosphate dehydrogenase,
glutathione, nitric oxide and circulating obestatin concentration. However,
these deleterious effects except food intake were attenuated when
supplemented with melatonin. Conclusion Taken together, the present results indicate that HFD exposure causes
adipose-hepatic metabolic disturbance in obese animals, which are
accompanied by oxidative stress and inflammation. In addition, the present
results suggest that melatonin supplementation attenuates adipose-hepatic
metabolic dysfunction, accompanying obesity by suppression of oxidative
stress/inflammation-dependent mechanism and increasing circulating
obestatin.
Collapse
Affiliation(s)
- Mary J. Obayemi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Christopher O. Akintayo
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Adesola A. Oniyide
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Ayodeji Aturamu
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Olabimpe C. Badejogbin
- Department of Physiology, Benjamin Carson School of Medicine, Babcock
University, Ilishan-Remo, Nigeria
| | - Chukwubueze L. Atuma
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Azeezat O. Saidi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Hadiza Mahmud
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
| | - Kehinde S. Olaniyi
- Department of Physiology, College of Medicine and Health Sciences, Afe
Babalola University, Ado-Ekiti, Nigeria
- * E-mail: ,
| |
Collapse
|
11
|
The expression and clinical significance of GPR39 in colon cancer. Ir J Med Sci 2021; 191:1577-1585. [PMID: 34586565 DOI: 10.1007/s11845-021-02792-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/21/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Colorectal cancer is the third most common cancer and requires more prognostic biomarkers for precise treatment. GPR39 is a GPCR which can interact with Zn and modulate the colonocytes' survival. The clinical significance of GPR39 in colon cancer has never been reported. MATERIALS In our study, we compared GPR39 expression between colon cancers and tumor-adjacent tissues by retrieving TCGA data and detected the expression of GPR39 in colon cancers with qPCR and immunohistochemistry. The clinical significance of GPR39 was evaluated by analyzing the correlations with clinicopathological factors with the chi-square test. The prognostic significance of GPR39 was estimated with univariate and multivariate analyses. The expression of several other biomarkers including PPARG, EPCAM, and PD-L1 was investigated by re-analyzing TCGA data, qPCR, and IHC. The prognostic value of PPARG, EPCAM, and PD-L1 was also estimated with univariate analysis. RESULTS In both TCGA database and our 15 colon cancer pairs, GPR39 expression was significantly upregulated in colon cancer tissues. GPR39 was an independent prognostic biomarker in colon cancer for poor prognosis. With TCGA data re-analysis, qPCR, and IHC, we showed that GPR39 expression was significantly correlated with the expression of EPCAM and PD-L1, but not PPARG. EPCAM and PD-L1 were also unfavorable prognostic biomarkers of colon cancer. CONCLUSIONS GPR39 was upregulated in colon cancer tissues compared with tumor-adjacent tissues. GPR39 was an independent prognostic biomarker in colon cancer for poor prognosis. EPCAM and PD-L1 were substantially associated with GPR39 expression, and they were also identified as prognostic biomarkers in colon cancers.
Collapse
|
12
|
Xu Y, Barnes AP, Alkayed NJ. Role of GPR39 in Neurovascular Homeostasis and Disease. Int J Mol Sci 2021; 22:8200. [PMID: 34360964 PMCID: PMC8346997 DOI: 10.3390/ijms22158200] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/26/2022] Open
Abstract
GPR39, a member of the ghrelin family of G protein-coupled receptors, is zinc-responsive and contributes to the regulation of diverse neurovascular and neurologic functions. Accumulating evidence suggests a role as a homeostatic regulator of neuronal excitability, vascular tone, and the immune response. We review GPR39 structure, function, and signaling, including constitutive activity and biased signaling, and summarize its expression pattern in the central nervous system. We further discuss its recognized role in neurovascular, neurological, and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yifan Xu
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Anthony P. Barnes
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Nabil J. Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health and Science University, Portland, OR 97239, USA;
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA;
| |
Collapse
|
13
|
Gastrointestinal Motility and Gut Hormone Secretion in response to Shenhuang Plaster in a Postoperative Ileus Rat Model. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8859579. [PMID: 34122608 PMCID: PMC8189788 DOI: 10.1155/2021/8859579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/28/2022]
Abstract
Postoperative ileus (POI), a gastrointestinal function disorder, is a complication that arises from surgery. Shenhuang plaster (SHP) application to the Shenque acupoint (CV8) to promote the recovery of gastrointestinal function has achieved definite curative effects in clinical settings; however, the underlying pharmacological mechanism remains unknown. In this study, we evaluated the effects of SHP using a Sprague Dawley rat POI model. Then, gastrointestinal transit in different rat groups was evaluated by the movement of fluorescein-labelled dextran. Ghrelin, obestatin, motilin (MTL), and vasoactive intestinal peptide (VIP) plasma concentrations were measured via a radioimmunoassay. The expression of the ghrelin and obestatin receptors (GHS-R1α and GPR39) in the intestinal muscularis of rats in different groups was comparatively identified via western blotting. The results indicated that SHP application improved gastrointestinal motility in POI model rats. SHP application significantly increased ghrelin concentration and the expression of its receptor and inhibited obestatin concentration and the expression of its receptor in blood. Further, ghrelin concentration and the capability of gastrointestinal transit were positively correlated. Simultaneously, SHP application also promoted the secretion of other gastrointestinal motility hormones, such as MTL and VIP. Hence, these results provide evidence that SHP can promote the recovery of gastrointestinal transmission in POI rat models through regulation of ghrelin and other intestinal hormones.
Collapse
|
14
|
Laitakari A, Liu L, Frimurer TM, Holst B. The Zinc-Sensing Receptor GPR39 in Physiology and as a Pharmacological Target. Int J Mol Sci 2021; 22:ijms22083872. [PMID: 33918078 PMCID: PMC8070507 DOI: 10.3390/ijms22083872] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/16/2022] Open
Abstract
The G-protein coupled receptor GPR39 is abundantly expressed in various tissues and can be activated by changes in extracellular Zn2+ in physiological concentrations. Previously, genetically modified rodent models have been able to shed some light on the physiological functions of GPR39, and more recently the utilization of novel synthetic agonists has led to the unraveling of several new functions in the variety of tissues GPR39 is expressed. Indeed, GPR39 seems to be involved in many important metabolic and endocrine functions, but also to play a part in inflammation, cardiovascular diseases, saliva secretion, bone formation, male fertility, addictive and depression disorders and cancer. These new discoveries offer opportunities for the development of novel therapeutic approaches against many diseases where efficient therapeutics are still lacking. This review focuses on Zn2+ as an endogenous ligand as well as on the novel synthetic agonists of GPR39, placing special emphasis on the recently discovered physiological functions and discusses their pharmacological potential.
Collapse
Affiliation(s)
- Anna Laitakari
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.L.); (L.L.); (T.M.F.)
| | - Lingzhi Liu
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.L.); (L.L.); (T.M.F.)
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
| | - Thomas M. Frimurer
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.L.); (L.L.); (T.M.F.)
| | - Birgitte Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark; (A.L.); (L.L.); (T.M.F.)
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
15
|
Siodłak D, Nowak G, Mlyniec K. Interaction between zinc, the GPR39 zinc receptor and the serotonergic system in depression. Brain Res Bull 2021; 170:146-154. [PMID: 33549699 DOI: 10.1016/j.brainresbull.2021.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022]
Abstract
Zinc signalling has a crucial impact on the proper functioning of the brain. Disturbances within the zincergic system may lead to neuropsychological disorders, including major depression. Studying this disease and designing effective treatment is hampered by its heterogeneous etiology and the diversified nature of the symptoms. Over the years, studies have shown that zinc deficiency and disturbances in the expression profile of the zinc receptor - GPR39 - might be a useful neurobiological indicator of a pathological state. Zinc levels and the zinc receptor are altered by classic antidepressant treatment, which indicates possible reciprocity between the monoaminergic system and zinc signalling. Disruptions in this specific interplay might be a cause of a pathological depressive state, and restoring balance and cooperation between those systems might be key to a successful form of pharmacotherapy. In this review, we aim to describe interactions between the serotonergic and zincergic systems and to highlight their significance in the pathophysiology and treatment of depression.
Collapse
Affiliation(s)
- Dominika Siodłak
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL, 30-688, Krakow, Poland
| | - Gabriel Nowak
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL, 30-688, Krakow, Poland; Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Katarzyna Mlyniec
- Department of Pharmacobiology, Jagiellonian University Medical College, Medyczna 9, PL, 30-688, Krakow, Poland.
| |
Collapse
|
16
|
Zhang L, Song J, Zang Z, Tang H, Li W, Lai S, Deng C. Adaptive evolution of GPR39 in diverse directions in vertebrates. Gen Comp Endocrinol 2020; 299:113610. [PMID: 32916170 DOI: 10.1016/j.ygcen.2020.113610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/27/2020] [Accepted: 08/30/2020] [Indexed: 02/08/2023]
Abstract
G protein-coupled receptors (GPCRs) play an important role in physiology and disease and represent productive drug targets. Orphan GPCRs, which have unknown endogenous ligands, are considered drug targets and consequently have attracted great interest in identifying their endogenous cognate ligands for deorphanization. However, additional studies have shown that GPCRs, including many orphan GPCRs, can constitutively activate G protein signaling in a ligand-independent manner. GPR39 is such an orphan GPCR with constitutive activity. Here, we performed a phylogenetic and selection analysis of GPR39 in vertebrates, and we found that GPR39 underwent positive selection in different branches of vertebrates. Using luciferase reporter assays, we demonstrated that human, frog and chicken GPR39 can constitutively activate Gq and G12 signaling pathways in a ligand-independent manner. Zebrafish GPR39 can constitutively activate Gs, Gq and G12 signaling pathways in a ligand-independent manner. We further found that the zebrafish-H2967.35 site is crucial for the activity of the Gs signaling pathway. In addition, our mutagenesis studies indicated that the positive selection sites of GPR39 from different species had important effects on the constitutive activity of the receptor. Our results revealed the adaptive evolution of GPR39 in diverse directions, which led to differences in constitutive activity.
Collapse
Affiliation(s)
- Lina Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Jingjing Song
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Zhuqing Zang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Huihao Tang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Wei Li
- Department of Dermatovenereology, Rare Disease Center, West China Hospital, Sichuan University, No. 37 Guo Xue Xiang Street, Chengdu, Sichuan 610041, China
| | - Shanshan Lai
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| | - Cheng Deng
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
17
|
Jiang Y, Li T, Wu Y, Xu H, Xie C, Dong Y, Zhong L, Wang Z, Zhao H, Zhou Y, Li J, Ji N, Zeng X, Feng X, Chen Q. GPR39 Overexpression in OSCC Promotes YAP-Sustained Malignant Progression. J Dent Res 2020; 99:949-958. [PMID: 32325008 DOI: 10.1177/0022034520915877] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The clinical outcome of oral squamous cell carcinoma (OSCC) has not improved in recent years, mainly due to the limited effective targeted therapy that has been applied. Recently, a transcriptional coactivator, YAP, has been shown to have a key regulatory role in malignant progression in multiple cancers, including OSCC. But pharmacologically targeting YAP or the Hippo pathway, which is the main signaling pathway regulating YAP, has been proven to be challenging. Therefore, uncovering YAP upstream regulators in cancer would identify novel therapeutic targets for treatment of YAP-sustained cancers. Here, we showed that YAP was overactivated in OSCC and that high YAP activity in patients with OSCC was associated with malignant progression and poor survival. We uncovered that GPR39 (a G protein-coupled receptor) was overexpressed in OSCC, that the expression level of GPR39 was correlated with the activity level of YAP, and that the high GPR39 expression was associated with malignant progression and poor survival in patients with OSCC. Moreover, we found that GPR39 regulated YAP through a Gαq/11-RhoA-dependent signaling pathway. Importantly, inhibition of GPR39 resulted in YAP-sustained OSCC growth inhibition. Our findings suggest that GPR39 is a potential therapeutic target for OSCC treatment with itself as a biomarker.
Collapse
Affiliation(s)
- Y Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - T Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - C Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Dong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Zhong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - N Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - X Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Nishida K, Hasegawa A, Yamasaki S, Uchida R, Ohashi W, Kurashima Y, Kunisawa J, Kimura S, Iwanaga T, Watarai H, Hase K, Ogura H, Nakayama M, Kashiwakura JI, Okayama Y, Kubo M, Ohara O, Kiyono H, Koseki H, Murakami M, Hirano T. Mast cells play role in wound healing through the ZnT2/GPR39/IL-6 axis. Sci Rep 2019; 9:10842. [PMID: 31346193 PMCID: PMC6658492 DOI: 10.1038/s41598-019-47132-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 07/11/2019] [Indexed: 01/08/2023] Open
Abstract
Zinc (Zn) is an essential nutrient and its deficiency causes immunodeficiency and skin disorders. Various cells including mast cells release Zn-containing granules when activated; however, the biological role of the released Zn is currently unclear. Here we report our findings that Zn transporter ZnT2 is required for the release of Zn from mast cells. In addition, we found that Zn and mast cells induce IL-6 production from inflammatory cells such as skin fibroblasts and promote wound healing, a process that involves inflammation. Zn induces the production of a variety of pro-inflammatory cytokines including IL-6 through signaling pathways mediated by the Zn receptor GPR39. Consistent with these findings, wound healing was impaired in mice lacking IL-6 or GPR39. Thus, our results show that Zn and mast cells play a critical role in wound healing through activation of the GPR39/IL-6 signaling axis.
Collapse
Affiliation(s)
- Keigo Nishida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki-cho, Suzuka, Mie, 513-8670, Japan. .,Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.
| | - Aiko Hasegawa
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Department of Pediatrics, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-8621, Japan
| | - Satoru Yamasaki
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Ryota Uchida
- Laboratory of Immune Regulation, Graduate School of Pharmaceutical Sciences, Suzuka University of Medical Science, 3500-3 Minamitamagaki-cho, Suzuka, Mie, 513-8670, Japan
| | - Wakana Ohashi
- Laboratory for Homeostatic Network, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Department of Molecular and Medical Pharmacology, Graduate School of Medicine and Pharmaceutical Sciences for Research, University of Toyama, Toyama, 930-0194, Japan
| | - Yosuke Kurashima
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, the Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Institute for Global Prominent Research, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California San Diego, 9500 Gilman Dr. MC 0063, San Diego, CA, 92093-0063, United States.,Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi Saito, Ibaraki, Osaka, 567-0085, Japan.,Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Shunsuke Kimura
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan.,Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo, 060-8638, Japan
| | - Hiroshi Watarai
- Department of Immunology and Stem Cell Biology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo, 105-8512, Japan.,International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo (IMSUT), 108-8639, Tokyo, Japan
| | - Hideki Ogura
- Department of Microbiology, Hyogo College of Medicine 1-1, Mukogawa-cho, Nishinomiya, 663-8501, Japan
| | - Manabu Nakayama
- Laboratory of Medical Omics Research, Department of Frontier Research and Development, Kazusa DNA Research Institute,2-6-7 Kazusa-Kamatari, Kisarazu, Chiba, 292-0818, Japan
| | - Jun-Ichi Kashiwakura
- Laboratory of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Yoshimichi Okayama
- Allergy and Immunology Project Team, Center for Allergy, Center for Medical Education, Nihon University School of Medicine, 30-1 Oyaguchi Kamicho Itabashi-Ku, Tokyo, 173-8610, Japan
| | - Masato Kubo
- Laboratory for Cytokine Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan.,Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, 2669 Yamazaki, Noda-shi, Chiba, 278-0022, Japan
| | - Osamu Ohara
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, the Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, Japan.,Institute for Global Prominent Research, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba, 260-8670, Japan.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy and Vaccines (CU-UCSD cMAV), University of California San Diego, 9500 Gilman Dr. MC 0063, San Diego, CA, 92093-0063, United States.,Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi, Yokohama, Kanagawa, 230-0045, Japan
| | - Masaaki Murakami
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, 060-815, Japan
| | - Toshio Hirano
- Headquarters, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
19
|
Bao LZ, Shen M, Qudirat H, Shi JB, Su T, Song JW, Wang ZK, Zhao XX, Jing Q, Zheng X, Guo ZF. Obestatin ameliorates water retention in chronic heart failure by downregulating renal aquaporin 2 through GPR39, V2R and PPARG signaling. Life Sci 2019; 231:116493. [PMID: 31153818 DOI: 10.1016/j.lfs.2019.05.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/16/2019] [Accepted: 05/19/2019] [Indexed: 12/28/2022]
Abstract
AIMS Obestatin regulates water metabolism by inhibiting arginine vasopressin (AVP) release and upregulated obestatin has been detected in patients with chronic heart failure (CHF). However, the significance of obestatin in CHF, particularly with regard to water retention and aquaporin 2 (AQP2) expression, remains unknown. MAIN METHODS Using a CHF rat model, the effects of 2-week exogenous obestatin administration were evaluated. Expression of AQP2 was evaluated by immunoblotting, immunohistochemical staining, and quantitative real-time PCR (qPCR) in CHF rat model and mouse inner medullary collecting duct (mIMCD) 3 cell line. Moreover, the influence of obestatin on the genetic transcription profile in mIMCD3 cells was evaluated by microarray, and the potential regulatory mechanisms of obestatin on AQP2 were evaluated by RNA silencing of vasopressin receptor 2 (V2R), peroxisome proliferator-activated receptor gamma (PPARG), and G protein-coupled receptor 39 (GPR39). KEY FINDINGS Obestatin increased urinary output and improved expression of CHF biomarker without significantly altering cardiac function, plasma electrolyte concentrations, or the plasma AVP concentration. AQP2 expression was significantly reduced. The results of microarray analyses and qPCR indicated that mRNA levels of Aqp2, Pparg, and V2r were significantly decreased. Inhibition of V2r and Pparg mRNA further reduced the expression of AQP2, while the inhibitory efficacy of obestatin on AQP2 was significantly offset after Gpr39 knockdown. SIGNIFICANCE Long-term treatment with obestatin improves water retention in CHF by increasing urinary output through downregulation of AQP2 expression in renal IMCD cells. These effects may be at least partially mediated by regulation of GPR39, V2R and PPARG signaling.
Collapse
Affiliation(s)
- Li-Zhi Bao
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Ming Shen
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Hannisa Qudirat
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jian-Bo Shi
- Department of Cardiology, HongKou Branch of Changhai Hospital of PLA, Shanghai 200081, China
| | - Ting Su
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jing-Wen Song
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zhong-Kai Wang
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xian-Xian Zhao
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qing Jing
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Xing Zheng
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Zhi-Fu Guo
- Department of Cardiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
20
|
Cuzon Carlson VC, Ford MM, Carlson TL, Lomniczi A, Grant KA, Ferguson B, Cervera-Juanes RP. Modulation of Gpr39, a G-protein coupled receptor associated with alcohol use in non-human primates, curbs ethanol intake in mice. Neuropsychopharmacology 2019; 44:1103-1113. [PMID: 30610192 PMCID: PMC6461847 DOI: 10.1038/s41386-018-0308-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 12/13/2018] [Accepted: 12/26/2018] [Indexed: 12/17/2022]
Abstract
Alcohol use disorder (AUD) is a chronic condition with devastating health and socioeconomic effects. Still, pharmacotherapies to treat AUD are scarce. In a prior study aimed at identifying novel AUD therapeutic targets, we investigated the DNA methylome of the nucleus accumbens core (NAcc) of rhesus macaques after chronic alcohol use. The G-protein coupled receptor 39 (GPR39) gene was hypermethylated and its expression downregulated in heavy alcohol drinking macaques. GPR39 encodes a Zn2+-binding metabotropic receptor known to modulate excitatory and inhibitory neurotransmission, the balance of which is altered in AUD. These prior findings suggest that a GPR39 agonist would reduce alcohol intake. Using a drinking-in-the-dark two bottle choice (DID-2BC) model, we showed that an acute 7.5 mg/kg dose of the GPR39 agonist, TC-G 1008, reduced ethanol intake in mice without affecting total fluid intake, locomotor activity or saccharin preference. Furthermore, repeated doses of the agonist prevented ethanol escalation in an intermittent access 2BC paradigm (IA-2BC). This effect was reversible, as ethanol escalation followed agonist "wash out". As observed during the DID-2BC study, a subsequent acute agonist challenge during the IA-2BC procedure reduced ethanol intake by ~47%. Finally, Gpr39 activation was associated with changes in Gpr39 and Bdnf expression, and in glutamate release in the NAcc. Together, our findings suggest that GPR39 is a promising target for the development of prevention and treatment therapies for AUD.
Collapse
Affiliation(s)
- Verginia C Cuzon Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Timothy L Carlson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
| | - Alejandro Lomniczi
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA
| | - Kathleen A Grant
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | - Betsy Ferguson
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA
- Department of Molecular and Medical Genetics, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Rita P Cervera-Juanes
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Sciences University, Beaverton, Oregon, USA.
- Division of Genetics, Oregon National Primate Research, Oregon Health and Sciences University, Beaverton, Oregon, USA.
| |
Collapse
|
21
|
Stempniewicz A, Ceranowicz P, Warzecha Z. Potential Therapeutic Effects of Gut Hormones, Ghrelin and Obestatin in Oral Mucositis. Int J Mol Sci 2019; 20:ijms20071534. [PMID: 30934722 PMCID: PMC6479885 DOI: 10.3390/ijms20071534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy and/or head and neck radiotherapy are frequently associated with oral mucositis. Oral pain, odynophagia and dysphagia, opioid use, weight loss, dehydration, systemic infection, hospitalization and introduction of a feeding tube should be mentioned as the main determinated effect of oral mucositis. Oral mucositis leads to a decreased quality of life and an increase in treatment costs. Moreover, oral mucositis is a life-threatening disease. In addition to its own direct life-threatening consequences, it can also lead to a reduced survival due to the discontinuation or dose reduction of anti-neoplasm therapy. There are numerous strategies for the prevention or treatment of oral mucositis; however, their effectiveness is limited and does not correspond to expectations. This review is focused on the ghrelin and obestatin as potentially useful candidates for the prevention and treatment of chemo- or/and radiotherapy-induced oral mucositis.
Collapse
Affiliation(s)
- Agnieszka Stempniewicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| | - Piotr Ceranowicz
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| | - Zygmunt Warzecha
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, Grzegórzecka 16 St., 31-531 Krakow, Poland.
| |
Collapse
|
22
|
|
23
|
Olaniru OE, Pingitore A, Giera S, Piao X, Castañera González R, Jones PM, Persaud SJ. The adhesion receptor GPR56 is activated by extracellular matrix collagen III to improve β-cell function. Cell Mol Life Sci 2018; 75:4007-4019. [PMID: 29855662 PMCID: PMC6182347 DOI: 10.1007/s00018-018-2846-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/11/2018] [Accepted: 05/24/2018] [Indexed: 12/19/2022]
Abstract
AIMS G-protein coupled receptor 56 (GPR56) is the most abundant islet-expressed G-protein coupled receptor, suggesting a potential role in islet function. This study evaluated islet expression of GPR56 and its endogenous ligand collagen III, and their effects on β-cell function. METHODS GPR56 and collagen III expression in mouse and human pancreas sections was determined by fluorescence immunohistochemistry. Effects of collagen III on β-cell proliferation, apoptosis, intracellular calcium ([Ca2+]i) and insulin secretion were determined by cellular BrdU incorporation, caspase 3/7 activities, microfluorimetry and radioimmunoassay, respectively. The role of GPR56 in islet vascularisation and innervation was evaluated by immunohistochemical staining for CD31 and TUJ1, respectively, in pancreases from wildtype (WT) and Gpr56-/- mice, and the requirement of GPR56 for normal glucose homeostasis was determined by glucose tolerance tests in WT and Gpr56-/- mice. RESULTS Immunostaining of mouse and human pancreases revealed that GPR56 was expressed by islet β-cells while collagen III was confined to the peri-islet basement membrane and islet capillaries. Collagen III protected β-cells from cytokine-induced apoptosis, triggered increases in [Ca2+]i and potentiated glucose-induced insulin secretion from WT islets but not from Gpr56-/- islets. Deletion of GPR56 did not affect glucose-induced insulin secretion in vitro and it did not impair glucose tolerance in adult mice. GPR56 was not required for normal islet vascularisation or innervation. CONCLUSION We have demonstrated that collagen III improves islet function by increasing insulin secretion and protecting against apoptosis. Our data suggest that collagen III may be effective in optimising islet function to improve islet transplantation outcomes, and GPR56 may be a target for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Oladapo E Olaniru
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Attilio Pingitore
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Stefanie Giera
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Ramón Castañera González
- Department of General Surgery, Rio Carrión Hospital, University Hospital Complex of Palencia, Palencia, Spain
| | - Peter M Jones
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
24
|
Zhu D, Su Y, Zheng Y, Fu B, Tang L, Qin YX. Zinc regulates vascular endothelial cell activity through zinc-sensing receptor ZnR/GPR39. Am J Physiol Cell Physiol 2018; 314:C404-C414. [PMID: 29351417 PMCID: PMC5966790 DOI: 10.1152/ajpcell.00279.2017] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 11/28/2017] [Accepted: 12/11/2017] [Indexed: 01/20/2023]
Abstract
Zn2+ is an essential element for cell survival/growth, and its deficiency is linked to many disorders. Extracellular Zn2+ concentration changes participate in modulating fundamental cellular processes such as proliferation, secretion, ion transport, and cell signal transduction in a mechanism that is not well understood. Here, we hypothesize that the Zn2+-sensing receptor ZnR/G protein-coupled receptor 39 (GPR39), found in tissues where dynamic Zn2+ homeostasis takes place, enables extracellular Zn2+ to trigger intracellular signaling pathways regulating key cell functions in vascular cells. Thus, we investigated how extracellular Zn2+ regulates cell viability, proliferation, motility, angiogenesis, vascular tone, and inflammation through ZnR/GPR39 in endothelial cells. Knockdown of GPR39 through siRNA largely abolished Zn2+-triggered cellular activity changes, Ca2+ responses, as well as the downstream activation of Gαq-PLC pathways. Extracellular Zn2+ promoted vascular cell survival/growth through activation of cAMP and Akt as well as overexpressing of platelet-derived growth factor-α receptor and vascular endothelial growth factor A. It also enhanced cell adhesion and mobility, endothelial tubule formation, and cytoskeletal reorganization. Such effects from extracellular Zn2+ were not observed in GPR39-/- endothelial cells. Zn2+ also regulated inflammation-related key molecules such as heme oxygenase-1, selectin L, IL-10, and platelet endothelial cell adhesion molecule 1, as well as vascular tone-related prostaglandin I2 synthase and nitric oxide synthase-3. In sum, extracellular Zn2+ regulates endothelial cell activity in a ZnR/GPR39-dependent manner and through the downstream Gαq-PLC pathways. Thus, ZnR/GPR39 may be a therapeutic target for regulating endothelial activity.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Biomedical Engineering, University of North Texas , Denton, Texas
| | - Yingchao Su
- Department of Biomedical Engineering, University of North Texas , Denton, Texas
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University , Beijing , China
| | - Bingmei Fu
- Department of Biomedical Engineering, The City College of the City University of New York , New York, New York
| | - Liping Tang
- Department of Bioengineering, University of Texas at Arlington , Arlington, Texas
| | - Yi-Xian Qin
- Department of Biomedical Engineering, State University of New York at Stony Brook , Stony Brook, New York
| |
Collapse
|
25
|
González-Mariscal L, Raya-Sandino A, González-González L, Hernández-Guzmán C. Relationship between G proteins coupled receptors and tight junctions. Tissue Barriers 2018; 6:e1414015. [PMID: 29420165 DOI: 10.1080/21688370.2017.1414015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tight junctions (TJs) are sites of cell-cell adhesion, constituted by a cytoplasmic plaque of molecules linked to integral proteins that form a network of strands around epithelial and endothelial cells at the uppermost portion of the lateral membrane. TJs maintain plasma membrane polarity and form channels and barriers that regulate the transit of ions and molecules through the paracellular pathway. This structure that regulates traffic between the external milieu and the organism is affected in numerous pathological conditions and constitutes an important target for therapeutic intervention. Here, we describe how a wide array of G protein-coupled receptors that are activated by diverse stimuli including light, ions, hormones, peptides, lipids, nucleotides and proteases, signal through heterotrimeric G proteins, arrestins and kinases to regulate TJs present in the blood-brain barrier, the blood-retinal barrier, renal tubular cells, keratinocytes, lung and colon, and the slit diaphragm of the glomerulus.
Collapse
Affiliation(s)
- Lorenza González-Mariscal
- a Department of Physiology , Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav) , Mexico City , Mexico
| | - Arturo Raya-Sandino
- a Department of Physiology , Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav) , Mexico City , Mexico
| | - Laura González-González
- a Department of Physiology , Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav) , Mexico City , Mexico
| | - Christian Hernández-Guzmán
- a Department of Physiology , Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav) , Mexico City , Mexico
| |
Collapse
|
26
|
Green BD, Grieve DJ. Biochemical properties and biological actions of obestatin and its relevence in type 2 diabetes. Peptides 2018; 100:249-259. [PMID: 29412827 DOI: 10.1016/j.peptides.2017.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/15/2022]
Abstract
Obestatin was initially discovered in rat stomach extract, and although it is principally produced in the gastric mucosa, it can be found throughout the gastrointestinal tract. This 23-amino acid C-terminally amidated peptide is derived from preproghrelin and has been ascribed a wide range of metabolic effects relevant to type 2 diabetes. Obestatin reportedly inhibits gastrointestinal motility, reduces food intake and lowers body weight and improves lipid metabolism. Furthermore, it appears to exert actions on the pancreatic β-cell, most notably increasing β-cell mass and upregulating genes associated with insulin production and β-cell regeneration, with relevance to type 2 diabetes. It is becoming evident that obestatin also exerts pleiotropic effects on the cardiovascular system, possibly modulating blood pressure, endothelial function and triggering cardioprotective mechanisms, which may be important in determining cardiovascular outcomes in type 2 diabetes. Furthermore, it seems that like other gut peptides obestatin has neuroprotective properties. This review examines the biochemical properties of the obestatin peptide (its structure, sequence, stability and distribution) and the candidate receptors through which it may act. It provides a balanced examination of the reported pancreatic and extrapancreatic actions of obestatin and evaluates its potential relevance with respect to diabetes therapy, together with discussion of direct evidence linking alterations in obestatin signalling with obesity/diabetes and other diseases.
Collapse
Affiliation(s)
- Brian D Green
- Institute for Global Food Security, School of Biological Sciences, Queen's University Belfast, Belfast, BT9 5BN, UK.
| | - David J Grieve
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7AE, UK
| |
Collapse
|
27
|
Hershfinkel M. The Zinc Sensing Receptor, ZnR/GPR39, in Health and Disease. Int J Mol Sci 2018; 19:ijms19020439. [PMID: 29389900 PMCID: PMC5855661 DOI: 10.3390/ijms19020439] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 01/24/2018] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
A distinct G-protein coupled receptor that senses changes in extracellular Zn2+, ZnR/GPR39, was found in cells from tissues in which Zn2+ plays a physiological role. Most prominently, ZnR/GPR39 activity was described in prostate cancer, skin keratinocytes, and colon epithelial cells, where zinc is essential for cell growth, wound closure, and barrier formation. ZnR/GPR39 activity was also described in neurons that are postsynaptic to vesicular Zn2+ release. Activation of ZnR/GPR39 triggers Gαq-dependent signaling and subsequent cellular pathways associated with cell growth and survival. Furthermore, ZnR/GPR39 was shown to regulate the activity of ion transport mechanisms that are essential for the physiological function of epithelial and neuronal cells. Thus, ZnR/GPR39 provides a unique target for therapeutically modifying the actions of zinc in a specific and selective manner.
Collapse
Affiliation(s)
- Michal Hershfinkel
- Department of Physiology and Cell Biology and The Zlotowski Center for Neuroscience, Faculty of Health Sciences, POB 653, Ben-Gurion Ave. Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.
| |
Collapse
|
28
|
Jovanovic M, Schmidt FN, Guterman-Ram G, Khayyeri H, Hiram-Bab S, Orenbuch A, Katchkovsky S, Aflalo A, Isaksson H, Busse B, Jähn K, Levaot N. Perturbed bone composition and integrity with disorganized osteoblast function in zinc receptor/Gpr39-deficient mice. FASEB J 2018; 32:2507-2518. [PMID: 29295862 DOI: 10.1096/fj.201700661rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Changes in bone matrix composition are frequently found with bone diseases and may be associated with increased fracture risk. Bone is rich in the trace element zinc. Zinc was established to play a significant role in the growth, development, and maintenance of healthy bones; however, the mechanisms underlying zinc effects on the integrity of the skeleton are poorly understood. Here, we show that the zinc receptor (ZnR)/Gpr39 is required for normal bone matrix deposition by osteoblasts. Initial analysis showed that Gpr39-deficient ( Gpr39-/-) mice had weaker bones as a result of altered bone composition. Fourier transform infrared spectroscopy analysis showed high mineral-to-matrix ratios in the bones of Gpr39-/- mice. Histologic analysis showed abnormally high numbers of active osteoblasts but normal osteoclast numbers on the surfaces of bones from Gpr39-/- mice. Furthermore, Gpr39-/- osteoblasts had disorganized matrix deposition in vitro with cultures exhibiting abnormally low collagen and high mineral contents, findings that demonstrate a cell-intrinsic role for ZnR/Gpr39 in these cells. We show that both collagen synthesis and deposition by Gpr39-/- osteoblasts are perturbed. Finally, the expression of the zinc transporter Zip13 and a disintegrin and metalloproteinase with thrombospondin motifs family of zinc-dependent metalloproteases that regulate collagen processing was downregulated in Gpr39-/- osteoblasts. Altogether, our results suggest that zinc sensing by ZnR/Gpr39 affects the expression levels of zinc-dependent enzymes in osteoblasts and regulates collagen processing and deposition.-Jovanovic, M., Schmidt, F. N., Guterman-Ram, G., Khayyeri, H., Hiram-Bab, S., Orenbuch, A., Katchkovsky, S., Aflalo, A., Isaksson, H., Busse, B., Jähn, K., Levaot, N. Perturbed bone composition and integrity with disorganized osteoblast function in zinc receptor/Gpr39-deficient mice.
Collapse
Affiliation(s)
- Milena Jovanovic
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Felix N Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gali Guterman-Ram
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hanifeh Khayyeri
- Department of Biomedical Engineering, Faculty of Engineering, Lund University, Lund, Sweden
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; and
| | - Ayelet Orenbuch
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Svetlana Katchkovsky
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anastasia Aflalo
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hanna Isaksson
- Department of Biomedical Engineering, Faculty of Engineering, Lund University, Lund, Sweden
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Jähn
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Noam Levaot
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Regenerative Medicine and Stem Cell Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
29
|
Sunuwar L, Medini M, Cohen L, Sekler I, Hershfinkel M. The zinc sensing receptor, ZnR/GPR39, triggers metabotropic calcium signalling in colonocytes and regulates occludin recovery in experimental colitis. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0420. [PMID: 27377730 DOI: 10.1098/rstb.2015.0420] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2016] [Indexed: 12/22/2022] Open
Abstract
Impaired epithelial barrier function is a hallmark of inflammatory bowel diseases, such as colitis, contributing to diarrhoea and perpetuating inflammation. We show that the zinc sensing receptor, ZnR/GPR39, triggers intracellular Ca(2+) signalling in colonocytes thereby inducing occludin expression. Moreover, ZnR/GPR39 is essential for epithelial barrier recovery in the dextran sodium sulfate (DSS) ulcerative colitis model. Loss of ZnR/GPR39 results in increased susceptibility to DSS-induced inflammation, owing to low expression of the tight junction protein occludin and impaired epithelial barrier. Recovery of wild-type (WT) mice from the DSS insult was faster than that of ZnR/GPR39 knockout (KO) mice. Enhanced recovery of the epithelial layer and increased crypt regeneration were observed in WT mice compared with ZnR/GPR39 KO, suggesting that ZnR/GPR39 is promoting epithelial barrier integrity following DSS insult. Indeed, cell proliferation and apical expression of occludin, following the DSS-induced epithelial erosion, were increased in WT tissue but not in ZnR/GPR39 KO tissue. Importantly, survival following DSS treatment was higher in WT mice compared with ZnR/GPR39 KO mice. Our results support a direct role for ZnR/GPR39 in promoting epithelial renewal and barrier function following DSS treatment, thereby affecting the severity of the disease. We suggest ZnR/GPR39 as a novel therapeutic target that can improve epithelial barrier function in colitis.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Michal Medini
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Limor Cohen
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| |
Collapse
|
30
|
Shimizu Y, Koyama R, Kawamoto T. Rho kinase-dependent desensitization of GPR39; a unique mechanism of GPCR downregulation. Biochem Pharmacol 2017; 140:105-114. [PMID: 28619258 DOI: 10.1016/j.bcp.2017.06.115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/09/2017] [Indexed: 12/14/2022]
Abstract
GPR39, a G-protein-coupled receptor activated by zinc, reportedly activates multiple intracellular signaling pathways via Gs, Gq, G12/13, and β-arrestin, but little is known about downregulation of the receptor upon its activation. To our knowledge, this is the first report on the mechanism of feedback regulation of GPR39 function determined in GPR39-expressing HEK293 cells (HEK293-GPR39) as a model cell system. In HEK293-GPR39 cells, GPR39-C3, which is a positive allosteric modulator, activated cAMP production (downstream of Gs), IP1 accumulation (downstream of Gq), SRF-RE-dependent transcription (downstream of G12/13), and β-arrestin recruitment. GPR39-C3 induced dose- and time-dependent loss of response in cAMP production by second challenge of the compound. This functional desensitization was blocked by the Rho kinase (ROCK) inhibitor, Y-27632, but not by Gq or Gs-pathway inhibitors or inhibition of β-arrestin recruitment. In the receptor localization assay, GPR39-C3 induced internalization of GFP-tagged GPR39. This internalization was also inhibited by Y-27632, which suggested that ROCK activation is critical for internalization and desensitization of GPR39. A novel biased GPR39 positive allosteric modulator, 5-[2-[(2,4-dichlorophenyl)methoxy]phenyl]-2,2-dimethyl-1,3,5,6-tetrahydrobenzo[a]phenanthridin-4-one (GSB-118), which activated cAMP responses and β-arrestin recruitment but showed no effect on SRF-RE-dependent transcription, did not induce desensitization. These results revealed a unique mechanism of desensitization of GPR39.
Collapse
Affiliation(s)
- Yuji Shimizu
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan.
| | - Ryokichi Koyama
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Tomohiro Kawamoto
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
31
|
Pradhan G, Wu CS, Han Lee J, Kanikarla P, Guo S, Yechoor VK, Samson SL, Sun Y. Obestatin stimulates glucose-induced insulin secretion through ghrelin receptor GHS-R. Sci Rep 2017; 7:979. [PMID: 28428639 PMCID: PMC5430520 DOI: 10.1038/s41598-017-00888-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 03/07/2017] [Indexed: 11/19/2022] Open
Abstract
Orexigenic hormone ghrelin and anorexic hormone obestatin are encoded by the same preproghrelin gene. While it is known that ghrelin inhibits glucose-stimulated insulin secretion (GSIS), the effect of obestatin on GSIS is unclear. Ghrelin’s effect is mediated by its receptor Growth Hormone Secretagogue Receptor (GHS-R), but the physiologically relevant receptor of obestatin remains debatable. Here we have investigated the effect of obestatin on GSIS in vitro, in vivo and ex vivo, and tested whether obestatin regulates insulin secretion through GHS-R. We found that under hyperglycemic condition, obestatin augments GSIS in rat insulinoma cells (INS-1) and in pancreatic islets from ghrelin−/− mice. Surprisingly, obestatin-induced GSIS was absent in β-cells in which GHS-R was suppressed. Obestatin-induced insulin secretion was abolished in the circulation of Ghsr−/− mice, and in pancreatic islets isolated from Ghsr−/− mice. We also found that obestatin-induced GSIS was attenuated in islets isolated from β-cell-specific Ghsr knockout MIP-Cre/ERT;Ghsrf/f mice. Our data collectively demonstrate that obestatin is a potent insulin secretagogue under hyperglycemic condition, and obestatin’s effect on insulin secretion is mediated by GHS-R in pancreatic β-cells. Our findings reveal an intriguing insight that obestatin and ghrelin have opposing effects on insulin secretion, and both are mediated through ghrelin receptor GHS-R.
Collapse
Affiliation(s)
- Geetali Pradhan
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Chia-Shan Wu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Jong Han Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,College of Pharmacy, Gachon University, Incheon, 21936, Korea
| | - Preeti Kanikarla
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Shaodong Guo
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA
| | - Vijay K Yechoor
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Susan L Samson
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yuxiang Sun
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA. .,Department of Nutrition and Food Science, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
32
|
Sunuwar L, Asraf H, Donowitz M, Sekler I, Hershfinkel M. The Zn 2+-sensing receptor, ZnR/GPR39, upregulates colonocytic Cl - absorption, via basolateral KCC1, and reduces fluid loss. Biochim Biophys Acta Mol Basis Dis 2017; 1863:947-960. [PMID: 28093242 PMCID: PMC5557417 DOI: 10.1016/j.bbadis.2017.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/13/2022]
Abstract
Administration of zinc, as a complement to oral rehydration solutions, effectively diminishes duration and severity of diarrhea, but it is not known whether it merely fulfills a nutritional deficiency, or if zinc has a direct role of regulating solute absorption. We show that Zn2+ acts via a specific receptor, ZnR/GPR39, to reduce fluid loss. Intestinal fluid secretion triggered by cholera toxin (CTx) was lower in WT mice compared to ZnR/GPR39 KO. In the absence of dietary Zn2+ we observed similar fluid accumulation in WT and ZnR/GPR39 KO mice, indicating that Zn2+ and ZnR/GPR39 are both required for a beneficial effect of Zn2+ in diarrhea. In primary colonocytes and in Caco-2 colonocytic cells, activation of ZnR/GPR39 enhanced Cl- transport, a critical factor in diarrhea, by upregulating K+/Cl- cotransporter (KCC1) activity. Importantly, we show basolateral expression of KCC1 in mouse and human colonocytes, thus identifying a novel Cl- absorption pathway. Finally, inhibition of KCC-dependent Cl- transport enhanced CTx-induced fluid loss. Altogether, our data indicate that Zn2+ acting via ZnR/GPR39 has a direct role in controlling Cl- absorption via upregulation of basolateral KCC1 in the colon. Moreover, colonocytic ZnR/GPR39 and KCC1 reduce water loss during diarrhea and may therefore serve as effective drug targets.
Collapse
Affiliation(s)
- Laxmi Sunuwar
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hila Asraf
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Mark Donowitz
- Department of Medicine, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
33
|
Khan MZ, He L. Neuro-psychopharmacological perspective of Orphan receptors of Rhodopsin (class A) family of G protein-coupled receptors. Psychopharmacology (Berl) 2017; 234:1181-1207. [PMID: 28289782 DOI: 10.1007/s00213-017-4586-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/27/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND In the central nervous system (CNS), G protein-coupled receptors (GPCRs) are the most fruitful targets for neuropsychopharmacological drug development. Rhodopsin (class A) is the most studied class of GPCR and includes orphan receptors for which the endogenous ligand is not known or is unclear. Characterization of orphan GPCRs has proven to be challenging, and the production pace of GPCR-based drugs has been incredibly slow. OBJECTIVE Determination of the functions of these receptors may provide unexpected insight into physiological and neuropathological processes. Advances in various methods and techniques to investigate orphan receptors including in situ hybridization and knockdown/knockout (KD/KO) showed extensive expression of these receptors in the mammalian brain and unmasked their physiological and neuropathological roles. Due to these rapid progress and development, orphan GPCRs are rising as a new and promising class of drug targets for neurodegenerative diseases and psychiatric disorders. CONCLUSION This review presents a neuropsychopharmacological perspective of 26 orphan receptors of rhodopsin (class A) family, namely GPR3, GPR6, GPR12, GPR17, GPR26, GPR35, GPR39, GPR48, GPR49, GPR50, GPR52, GPR55, GPR61, GPR62, GPR63, GPR68, GPR75, GPR78, GPR83, GPR84, GPR85, GPR88, GPR153, GPR162, GPR171, and TAAR6. We discussed the expression of these receptors in mammalian brain and their physiological roles. Furthermore, we have briefly highlighted their roles in neurodegenerative diseases and psychiatric disorders including Alzheimer's disease, Parkinson's disease, neuroinflammation, inflammatory pain, bipolar and schizophrenic disorders, epilepsy, anxiety, and depression.
Collapse
Affiliation(s)
- Muhammad Zahid Khan
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China.
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, No. 24 Tong Jia Xiang, Nanjing, Jiangsu Province, 210009, China
| |
Collapse
|
34
|
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
|
35
|
Fatty acid and mineral receptors as drug targets for gastrointestinal disorders. Future Med Chem 2017; 9:315-334. [DOI: 10.4155/fmc-2016-0205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nutrient-sensing receptors, including fatty acid receptors (FFA1–FFA4), Ca2+-sensing receptors and Zn2+-sensing receptors, are involved in several biological processes. These receptors are abundantly expressed in the GI tract, where they have been shown to play crucial roles in regulating GI function. This review provides an overview of the GI functions of fatty acid and mineral receptors, including the regulation of gastric and enteroendocrine functions, GI motility, ion transport and cell growth. Recently, several lines of evidence have implicated these receptors as promising therapeutic targets for the treatment of GI disorders, for example, inflammatory bowel disease, colorectal cancer, metabolic syndrome and diarrheal diseases. A future perspective on drug discovery research targeting these receptors is discussed.
Collapse
|
36
|
Model-Based Discovery of Synthetic Agonists for the Zn 2+-Sensing G-Protein-Coupled Receptor 39 (GPR39) Reveals Novel Biological Functions. J Med Chem 2017; 60:886-898. [PMID: 28045522 DOI: 10.1021/acs.jmedchem.6b00648] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The G-protein-coupled receptor 39 (GPR39) is a G-protein-coupled receptor activated by Zn2+. We used a homology model-based approach to identify small-molecule pharmacological tool compounds for the receptor. The method focused on a putative binding site in GPR39 for synthetic ligands and knowledge of ligand binding to other receptors with similar binding pockets to select iterative series of minilibraries. These libraries were cherry-picked from all commercially available synthetic compounds. A total of only 520 compounds were tested in vitro, making this method broadly applicable for tool compound development. The compounds of the initial library were inactive when tested alone, but lead compounds were identified using Zn2+ as an allosteric enhancer. Highly selective, highly potent Zn2+-independent GPR39 agonists were found in subsequent minilibraries. These agonists identified GPR39 as a novel regulator of gastric somatostatin secretion.
Collapse
|
37
|
Hassouna R, Labarthe A, Tolle V. Hypothalamic regulation of body growth and appetite by ghrelin-derived peptides during balanced nutrition or undernutrition. Mol Cell Endocrinol 2016; 438:42-51. [PMID: 27693419 DOI: 10.1016/j.mce.2016.09.027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 09/25/2016] [Accepted: 09/26/2016] [Indexed: 12/16/2022]
Abstract
Among the gastrointestinal hormones that regulate food intake and energy homeostasis, ghrelin plays a unique role as the first one identified to increases appetite and stimulate GH secretion. This review highlights the latest mechanism by which ghrelin modulates body growth, appetite and energy metabolism by exploring pharmacological actions of the hormone and consequences of genetic or pharmacological blockade of the ghrelin/GHS-R (Growth Hormone Secretagogue Receptor) system on physiological responses in specific nutritional situations. Within the hypothalamus, novel mechanisms of action of this hormone involve its interaction with other ghrelin-derived peptides, such as desacyl ghrelin and obestatin, which are thought to act as functional ghrelin antagonists, and possible modulation of the GHS-R with other G-protein coupled receptors. During chronic undernutrition such as anorexia nervosa, variations of ghrelin-derived peptides may be an adaptative metabolic response to maintain normal glycemic control. Interestingly, some of ghrelin's metabolic actions are thought to be relayed through modulation of GH, an anabolic and hyperglycemic agent.
Collapse
Affiliation(s)
- Rim Hassouna
- UMR-S 894 INSERM, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, 2 ter rue d'Alésia, 75014, Paris, France; Naomi Berrie Diabetes Center, Department of Pediatrics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Alexandra Labarthe
- UMR-S 894 INSERM, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, 2 ter rue d'Alésia, 75014, Paris, France
| | - Virginie Tolle
- UMR-S 894 INSERM, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, Sorbonne Paris Cité, 2 ter rue d'Alésia, 75014, Paris, France.
| |
Collapse
|
38
|
Abramovitch-Dahan C, Asraf H, Bogdanovic M, Sekler I, Bush AI, Hershfinkel M. Amyloid β attenuates metabotropic zinc sensing receptor, mZnR/GPR39, dependent Ca2+
, ERK1/2 and Clusterin signaling in neurons. J Neurochem 2016; 139:221-233. [DOI: 10.1111/jnc.13760] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Chen Abramovitch-Dahan
- Department of Physiology and Cell Biology; Faculty of Health Sciences; Ben-Gurion University of the Negev; Beer-Sheva Israel
| | - Hila Asraf
- Department of Physiology and Cell Biology; Faculty of Health Sciences; Ben-Gurion University of the Negev; Beer-Sheva Israel
| | - Milos Bogdanovic
- Department of Physiology and Cell Biology; Faculty of Health Sciences; Ben-Gurion University of the Negev; Beer-Sheva Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology; Faculty of Health Sciences; Ben-Gurion University of the Negev; Beer-Sheva Israel
| | - Ashley I. Bush
- Florey Institute of Neuroscience and Mental Health; University of Melbourne; Parkville Victoria Australia
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology; Faculty of Health Sciences; Ben-Gurion University of the Negev; Beer-Sheva Israel
| |
Collapse
|
39
|
Cowan E, Burch KJ, Green BD, Grieve DJ. Obestatin as a key regulator of metabolism and cardiovascular function with emerging therapeutic potential for diabetes. Br J Pharmacol 2016; 173:2165-81. [PMID: 27111465 DOI: 10.1111/bph.13502] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/05/2016] [Accepted: 04/15/2016] [Indexed: 01/01/2023] Open
Abstract
Obestatin is a 23-amino acid C-terminally amidated gastrointestinal peptide derived from preproghrelin and which forms an α helix. Although obestatin has a short biological half-life and is rapidly degraded, it is proposed to exert wide-ranging pathophysiological actions. Whilst the precise nature of many of its effects is unclear, accumulating evidence supports positive actions on both metabolism and cardiovascular function. For example, obestatin has been reported to inhibit food and water intake, body weight gain and gastrointestinal motility and also to mediate promotion of cell survival and prevention of apoptosis. Obestatin-induced increases in beta cell mass, enhanced adipogenesis and improved lipid metabolism have been noted along with up-regulation of genes associated with beta cell regeneration, insulin production and adipogenesis. Furthermore, human circulating obestatin levels generally demonstrate an inverse association with obesity and diabetes, whilst the peptide has been shown to confer protective metabolic effects in experimental diabetes, suggesting that it may hold therapeutic potential in this setting. Obestatin also appears to be involved in blood pressure regulation and to exert beneficial effects on endothelial function, with experimental studies indicating that it may also promote cardioprotective actions against, for example, ischaemia-reperfusion injury. This review will present a critical appraisal of the expanding obestatin research area and discuss the emerging therapeutic potential of this peptide for both metabolic and cardiovascular complications of diabetes.
Collapse
Affiliation(s)
- Elaine Cowan
- Queen's University Belfast, Institute for Global Food Security, School of Biological Sciences, Belfast, UK
| | - Kerry J Burch
- Queen's University Belfast, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| | - Brian D Green
- Queen's University Belfast, Institute for Global Food Security, School of Biological Sciences, Belfast, UK
| | - David J Grieve
- Queen's University Belfast, Wellcome-Wolfson Institute for Experimental Medicine, Belfast, UK
| |
Collapse
|
40
|
Moran BM, Abdel-Wahab YHA, Vasu S, Flatt PR, McKillop AM. GPR39 receptors and actions of trace metals on pancreatic beta cell function and glucose homoeostasis. Acta Diabetol 2016; 53:279-93. [PMID: 26112416 DOI: 10.1007/s00592-015-0781-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 05/29/2015] [Indexed: 10/23/2022]
Abstract
AIMS G-protein-coupled receptor 39 (GPR39) has been implicated in glucose homoeostasis, appetite control and gastrointestinal tract function. METHODS This study used clonal BRIN-BD11 cells and mouse pancreatic islets to assess the insulin-releasing actions of trace metals believed to act via GPR39, and the second messenger pathways involved in mediating their effects. Micromolar concentrations of Zn(2+), Cu(2+), Ni(2+) and Co(2+) were examined under normoglycaemic and hyperglycaemic conditions. Mechanistic studies investigated changes of intracellular Ca(2+), cAMP generation and assessment of cytotoxicity by LDH release. Cellular localisation of GPR39 was determined by double immunohistochemical staining. RESULTS All trace metals (7.8-500 µmol/l) stimulated insulin release with Cu(2+) being the most potent in isolated islets, with an EC50 value of 87 μmol/l. Zn(2+) was the most selective with an EC50 value of 125 μmol/l. Enhancement of insulin secretion was also observed with Ni(2+) (179 μmol/l) and Co(2+) (190 μmol/l). These insulin-releasing effects were confirmed using clonal BRIN-BD11 cells which exhibited enhanced intracellular Ca(2+) (p < 0.05-p < 0.001) and cAMP generation (p < 0.05-p < 0.001) in response to trace metals. Oral administration of Zn(2+), Ni(2+) and Cu(2+) (50 µmol/kg together with 18 mmol/kg glucose) decreased the glycaemic excursion (p < 0.05-p < 0.01) and augmented insulin secretion (p < 0.05-p < 0.01) in NIH Swiss mice. CONCLUSIONS This study has demonstrated the presence of GPR39 and the insulinotropic actions of trace metals on BRIN-BD11 cells and pancreatic beta cells, together with their antihyperglycaemic actions in vivo. These data suggest that development of agonists capable of specifically activating GPR39 may be a useful new therapeutic approach for diabetes management.
Collapse
Affiliation(s)
- Brian M Moran
- Biomedical Sciences Research Institute, SAAD Centre for Pharmacy and Diabetes, University of Ulster, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Yasser H A Abdel-Wahab
- Biomedical Sciences Research Institute, SAAD Centre for Pharmacy and Diabetes, University of Ulster, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Srividya Vasu
- Biomedical Sciences Research Institute, SAAD Centre for Pharmacy and Diabetes, University of Ulster, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Peter R Flatt
- Biomedical Sciences Research Institute, SAAD Centre for Pharmacy and Diabetes, University of Ulster, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, UK
| | - Aine M McKillop
- Biomedical Sciences Research Institute, SAAD Centre for Pharmacy and Diabetes, University of Ulster, Cromore Road, Coleraine, BT52 1SA, Northern Ireland, UK.
| |
Collapse
|
41
|
Hashimoto A, Nakagawa M, Tsujimura N, Miyazaki S, Kizu K, Goto T, Komatsu Y, Matsunaga A, Shirakawa H, Narita H, Kambe T, Komai M. Properties of Zip4 accumulation during zinc deficiency and its usefulness to evaluate zinc status: a study of the effects of zinc deficiency during lactation. Am J Physiol Regul Integr Comp Physiol 2016; 310:R459-68. [DOI: 10.1152/ajpregu.00439.2015] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/18/2015] [Indexed: 11/22/2022]
Abstract
Systemic and cellular zinc homeostasis is elaborately controlled by ZIP and ZnT zinc transporters. Therefore, detailed characterization of their expression properties is of importance. Of these transporter proteins, Zip4 functions as the primarily important transporter to control systemic zinc homeostasis because of its indispensable function of zinc absorption in the small intestine. In this study, we closely investigated Zip4 protein accumulation in the rat small intestine in response to zinc status using an anti-Zip4 monoclonal antibody that we generated and contrasted this with the zinc-responsive activity of the membrane-bound alkaline phosphatase (ALP). We found that Zip4 accumulation is more rapid in response to zinc deficiency than previously thought. Accumulation increased in the jejunum as early as 1 day following a zinc-deficient diet. In the small intestine, Zip4 protein expression was higher in the jejunum than in the duodenum and was accompanied by reduction of ALP activity, suggesting that the jejunum can become zinc deficient more easily. Furthermore, by monitoring Zip4 accumulation levels and ALP activity in the duodenum and jejunum, we reasserted that zinc deficiency during lactation may transiently alter plasma glucose levels in the offspring in a sex-specific manner, without affecting homeostatic control of zinc metabolism. This confirms that zinc nutrition during lactation is extremely important for the health of the offspring. These results reveal that rapid Zip4 accumulation provides a significant conceptual advance in understanding the molecular basis of systemic zinc homeostatic control, and that properties of Zip4 protein accumulation are useful to evaluate zinc status closely.
Collapse
Affiliation(s)
- Ayako Hashimoto
- The Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Miki Nakagawa
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Natsuki Tsujimura
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Shiho Miyazaki
- The Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kumiko Kizu
- Department of Life and Living, Osaka Seikei College, Osaka, Japan; and
| | - Tomoko Goto
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Yusuke Komatsu
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Ayu Matsunaga
- Department of Food Science, Kyoto Women's University, Kyoto, Japan
| | - Hitoshi Shirakawa
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| | - Hiroshi Narita
- Department of Food Science, Kyoto Women's University, Kyoto, Japan
| | - Taiho Kambe
- The Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Michio Komai
- Department of Science of Food Function and Health, Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
| |
Collapse
|
42
|
Fjellström O, Larsson N, Yasuda SI, Tsuchida T, Oguma T, Marley A, Wennberg-Huldt C, Hovdal D, Fukuda H, Yoneyama Y, Sasaki K, Johansson A, Lundqvist S, Brengdahl J, Isaacs RJ, Brown D, Geschwindner S, Benthem L, Priest C, Turnbull A. Novel Zn2+ Modulated GPR39 Receptor Agonists Do Not Drive Acute Insulin Secretion in Rodents. PLoS One 2015; 10:e0145849. [PMID: 26720709 PMCID: PMC4697807 DOI: 10.1371/journal.pone.0145849] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/09/2015] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes (T2D) occurs when there is insufficient insulin release to control blood glucose, due to insulin resistance and impaired β-cell function. The GPR39 receptor is expressed in metabolic tissues including pancreatic β-cells and has been proposed as a T2D target. Specifically, GPR39 agonists might improve β-cell function leading to more adequate and sustained insulin release and glucose control. The present study aimed to test the hypothesis that GPR39 agonism would improve glucose stimulated insulin secretion in vivo. A high throughput screen, followed by a medicinal chemistry program, identified three novel potent Zn2+ modulated GPR39 agonists. These agonists were evaluated in acute rodent glucose tolerance tests. The results showed a lack of glucose lowering and insulinotropic effects not only in lean mice, but also in diet-induced obese (DIO) mice and Zucker fatty rats. It is concluded that Zn2+ modulated GPR39 agonists do not acutely stimulate insulin release in rodents.
Collapse
Affiliation(s)
- Ola Fjellström
- Medicinal Chemistry CVMD iMed, AstraZeneca R&D Gothenburg, Mölndal, Sweden
- * E-mail:
| | - Niklas Larsson
- Discovery Sciences, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Shin-ichiro Yasuda
- Pharmacology Research Laboratories II, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Takuma Tsuchida
- Pharmacology Research Laboratories II, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Takahiro Oguma
- Pharmacology Research Laboratories II, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Anna Marley
- Discovery Sciences, AstraZeneca R&D, Mereside, United Kingdom
| | | | - Daniel Hovdal
- DMPK CVMD iMed, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Hajime Fukuda
- DMPK Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Yukimi Yoneyama
- DMPK Research Laboratories, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Kazuyo Sasaki
- Pharmacology Research Laboratories II, Mitsubishi Tanabe Pharma Corporation, Kawagishi, Toda-shi, Saitama, Japan
| | - Anders Johansson
- Medicinal Chemistry CVMD iMed, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Sara Lundqvist
- Discovery Sciences, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Johan Brengdahl
- Discovery Sciences, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Richard J. Isaacs
- Molecular Sensing, Inc., Nashville, Tennessee, United States of America
| | - Daniel Brown
- Molecular Sensing, Inc., Nashville, Tennessee, United States of America
| | | | | | - Claire Priest
- Discovery Sciences, AstraZeneca R&D, Mereside, United Kingdom
| | | |
Collapse
|
43
|
Tena-Campos M, Ramon E, Borroto-Escuela DO, Fuxe K, Garriga P. The zinc binding receptor GPR39 interacts with 5-HT1A and GalR1 to form dynamic heteroreceptor complexes with signaling diversity. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2585-92. [PMID: 26365466 DOI: 10.1016/j.bbadis.2015.09.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 09/02/2015] [Accepted: 09/09/2015] [Indexed: 01/20/2023]
Abstract
GPR39 is a class A G protein-coupled receptor involved in zinc binding and glucose homeostasis regulation, among other physiological processes. GPR39 was originally thought to be the receptor for obestatin peptide but this view has been challenged. However, activation of this receptor by zinc has been clearly established. Recent studies suggest that low GPR39 expression, due to deficient zinc levels, is involved in major depressive disorder. We have previously reported that zinc can alter receptor-receptor interactions and favor specific receptor interactions. In order to unravel the effect of zinc on specific G protein-coupled receptor association processes, we have performed FRET and co-immunopurification studies with GPR39 and 5-HT1A and GalR1 which have been shown to dimerize. Our results suggest that zinc can modulate the formation of specific 5-HT1A-GPR39 and GalR1-5-HT1A-GPR39 heteroreceptor complexes under our experimental conditions. We have analyzed the differences in signaling between the mono-homomeric receptors 5-HT1A, GalR1 and GPR39 and the heteroreceptor complexes between them Our results show that the GPR39-5-HT1A heterocomplex has additive functionalities when compared to the monomeric-homomeric receptors upon receptor activation. In addition, the heterocomplex including also GalR1 shows a different behavior, upon exposure to the same agonists. Furthermore, these processes appear to be regulated by zinc. These findings provide a rationale for the antidepressive effect widely described for zinc because pro-depressive heterocomplexes are predominant at low zinc concentration levels.
Collapse
Affiliation(s)
- Mercè Tena-Campos
- Departament d'Enginyeria Química, Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Universitat Politècnica de Catalunya, Edifici Gaia, Rambla de Sant Nebridi n° 22, 08222 Terrassa, Catalonia, Spain
| | - Eva Ramon
- Departament d'Enginyeria Química, Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Universitat Politècnica de Catalunya, Edifici Gaia, Rambla de Sant Nebridi n° 22, 08222 Terrassa, Catalonia, Spain
| | | | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Retzius väg 8, 17177 Stockhom, Sweden
| | - Pere Garriga
- Departament d'Enginyeria Química, Grup de Biotecnologia Molecular i Industrial, Centre de Biotecnologia Molecular, Universitat Politècnica de Catalunya, Edifici Gaia, Rambla de Sant Nebridi n° 22, 08222 Terrassa, Catalonia, Spain.
| |
Collapse
|
44
|
Gilad D, Shorer S, Ketzef M, Friedman A, Sekler I, Aizenman E, Hershfinkel M. Homeostatic regulation of KCC2 activity by the zinc receptor mZnR/GPR39 during seizures. Neurobiol Dis 2015; 81:4-13. [PMID: 25562657 PMCID: PMC4490144 DOI: 10.1016/j.nbd.2014.12.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 12/11/2014] [Accepted: 12/23/2014] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to investigate the role of the synaptic metabotropic zinc receptor mZnR/GPR39 in physiological adaptation to epileptic seizures. We previously demonstrated that synaptic activation of mZnR/GPR39 enhances inhibitory drive in the hippocampus by upregulating neuronal K(+)/Cl(-) co-transporter 2 (KCC2) activity. Here, we first show that mZnR/GPR39 knockout (KO) adult mice have dramatically enhanced susceptibility to seizures triggered by a single intraperitoneal injection of kainic acid, when compared to wild type (WT) littermates. Kainate also substantially enhances seizure-associated gamma oscillatory activity in juvenile mZnR/GPR39 KO hippocampal slices, a phenomenon that can be reproduced in WT tissue by extracellular Zn(2+) chelation. Importantly, kainate-induced synaptic Zn(2+) release enhances surface expression and transport activity of KCC2 in WT, but not mZnR/GPR39 KO hippocampal neurons. Kainate-dependent upregulation of KCC2 requires mZnR/GPR39 activation of the Gαq/phospholipase C/extracellular regulated kinase (ERK1/2) signaling cascade. We suggest that mZnR/GPR39-dependent upregulation of KCC2 activity provides homeostatic adaptation to an excitotoxic stimulus by increasing inhibition. As such, mZnR/GPR39 may provide a novel pharmacological target for dampening epileptic seizure activity.
Collapse
Affiliation(s)
- David Gilad
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Sharon Shorer
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Maya Ketzef
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Alon Friedman
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Israel Sekler
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel
| | - Elias Aizenman
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michal Hershfinkel
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Faculty of Health Sciences, Beer-Sheva, Israel.
| |
Collapse
|
45
|
Lemarié F, Beauchamp E, Legrand P, Rioux V. Revisiting the metabolism and physiological functions of caprylic acid (C8:0) with special focus on ghrelin octanoylation. Biochimie 2015; 120:40-8. [PMID: 26253695 DOI: 10.1016/j.biochi.2015.08.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/01/2015] [Indexed: 12/22/2022]
Abstract
Caprylic acid (octanoic acid, C8:0) belongs to the class of medium-chain saturated fatty acids (MCFAs). Dairy products and specific oils like coconut oil are natural sources of dietary C8:0 but higher intakes of this fatty acid can be provided with MCT (Medium-Chain Triglycerides) oil that consists in 75% of C8:0. MCFAs have physical and metabolic properties that are distinct from those of long-chain saturated fatty acids (LCFAs ≥ 12 carbons). Beneficial physiological effects of dietary C8:0 have been studied for a long time and MCT oil has been used as a special energy source for patients suffering from pancreatic insufficiency, impaired lymphatic chylomicron transport and fat malabsorption. More recently, caprylic acid was also shown to acylate ghrelin, the only known peptide hormone with an orexigenic effect. Through its covalent binding to the ghrelin peptide, caprylic acid exhibits an emerging and specific role in modulating physiological functions themselves regulated by octanoylated ghrelin. Dietary caprylic acid is therefore now suspected to provide the ghrelin O-acyltransferase (GOAT) enzyme with octanoyl-CoA co-substrates necessary for the acyl modification of ghrelin. This review tries to highlight the discrepancy between the formerly described beneficial effects of dietary MCFAs on body weight loss and the C8:0 newly reported effect on appetite stimulation via ghrelin octanoylation. The subsequent aim of this review is to demonstrate the relevance of carrying out further studies to better understand the physiological functions of this particular fatty acid.
Collapse
Affiliation(s)
- Fanny Lemarié
- Laboratoire de Biochimie-Nutrition Humaine, Agrocampus Ouest, INRA USC 1378, Rennes, France
| | - Erwan Beauchamp
- Laboratoire de Biochimie-Nutrition Humaine, Agrocampus Ouest, INRA USC 1378, Rennes, France
| | - Philippe Legrand
- Laboratoire de Biochimie-Nutrition Humaine, Agrocampus Ouest, INRA USC 1378, Rennes, France
| | - Vincent Rioux
- Laboratoire de Biochimie-Nutrition Humaine, Agrocampus Ouest, INRA USC 1378, Rennes, France.
| |
Collapse
|
46
|
GPR39 Zn(2+)-sensing receptor: a new target in antidepressant development? J Affect Disord 2015; 174:89-100. [PMID: 25490458 DOI: 10.1016/j.jad.2014.11.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/17/2014] [Accepted: 11/18/2014] [Indexed: 11/23/2022]
Abstract
Zinc is a trace element released from glutamatergic terminals, and modulates the pre- and postsynaptic areas, giving a diverse biological response. Zinc is a natural ligand that inhibits the N-methyl-d-aspartate (NMDA) receptor and regulates the excessive release of glutamate. Moreover, zinc exhibits an antidepressant-like profile, as demonstrated in both preclinical and clinical studies. Recent reports indicate that the GPR39 Zn(2+)-sensing receptor is an important target for zinc "transmission" (its activation modulates/induces diverse biochemical pathways involved in neuroprotection). Preclinical studies provide evidence that zinc deficiency leads to depressive-like behavior related to down-regulation of the GPR39 Zn(2+)-sensing receptor. Zinc binds to the GPR39 and triggers signals, leading to CRE-dependent gene transcription, resulting in increases in proteins such as brain-derived neurotrophic factor (BDNF), that plays a pivotal role in antidepressant action. Chronic administration of many antidepressants induces GPR39 up-regulation, which suggests that the Zn(2+)-sensing receptor may be considered as a new target for drug development in the field of depression.
Collapse
|
47
|
Zhao H, Qiao J, Zhang S, Zhang H, Lei X, Wang X, Deng Z, Ning L, Cao Y, Guo Y, Liu S, Duan E. GPR39 marks specific cells within the sebaceous gland and contributes to skin wound healing. Sci Rep 2015; 5:7913. [PMID: 25604641 PMCID: PMC4300488 DOI: 10.1038/srep07913] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 12/19/2014] [Indexed: 12/28/2022] Open
Abstract
G protein-coupled receptors (GPCRs) mediate multiple key biological processes in the body. The orphan receptor GPR39 has been reported to be involved in various pathophysiological events. However, the function of GPR39 in skin biology remains unknown. Using a genetically engineered mouse strain in which lacZ expression faithfully replaced endogenous Gpr39 expression, we discovered a unique expression pattern of Gpr39 in the sebaceous gland (SG). Using various methods, we confirmed that GPR39 marked a specific cell population at the opening of the SG and colocalised with the SG stem cell marker Blimp1. Further investigations showed that GPR39 was spatiotemporally expressed during skin wound repair. Although it was dispensable for skin development and homeostasis, GPR39 contributed positively to skin wound healing: its loss led to a delay in wound healing during the intermediate stage. The present study reveals a novel role of GPR39 in both dermatology and stem cell biology that has not been previously recognised.
Collapse
Affiliation(s)
- Huashan Zhao
- 1] State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China [2] University of Chinese Academy of Sciences, Beijing, China
| | - Jingqiao Qiao
- College of Animal Science and Technology, Beijing University of Agriculture, China
| | - Shoubing Zhang
- Department of Histology&Embryology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Huishan Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaohua Lei
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinyue Wang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhili Deng
- 1] State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China [2] University of Chinese Academy of Sciences, Beijing, China
| | - Lina Ning
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yujing Cao
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yong Guo
- College of Animal Science and Technology, Beijing University of Agriculture, China
| | - Shuang Liu
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Enkui Duan
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
48
|
Peukert S, Hughes R, Nunez J, He G, Yan Z, Jain R, Llamas L, Luchansky S, Carlson A, Liang G, Kunjathoor V, Pietropaolo M, Shapiro J, Castellana A, Wu X, Bose A. Discovery of 2-Pyridylpyrimidines as the First Orally Bioavailable GPR39 Agonists. ACS Med Chem Lett 2014; 5:1114-8. [PMID: 25313322 DOI: 10.1021/ml500240d] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 08/04/2014] [Indexed: 11/29/2022] Open
Abstract
The identification of highly potent and orally bioavailable GPR39 agonists is reported. Compound 1, found in a phenotypic screening campaign, was transformed into compound 2 with good activity on both the rat and human GPR39 receptor. This compound was further optimized to improve ligand efficiency and pharmacokinetic properties to yield GPR39 agonists for the potential oral treatment of type 2 diabetes. Thus, compound 3 is the first potent GPR39 agonist (EC50s ≤ 1 nM for human and rat receptor) that is orally bioavailable in mice and robustly induced acute GLP-1 levels.
Collapse
Affiliation(s)
- Stefan Peukert
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Richard Hughes
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jill Nunez
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Guo He
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Zhao Yan
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Rishi Jain
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Luis Llamas
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Sarah Luchansky
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Adam Carlson
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Guiqing Liang
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Vidya Kunjathoor
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Mike Pietropaolo
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Jeffrey Shapiro
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Anja Castellana
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Xiaoping Wu
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Avirup Bose
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
49
|
Protein kinase inhibitor β enhances the constitutive activity of G-protein-coupled zinc receptor GPR39. Biochem J 2014; 462:125-32. [PMID: 24869658 DOI: 10.1042/bj20131198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
GPR39 is a G-protein-coupled zinc receptor that protects against diverse effectors of cell death. Its protective activity is mediated via constitutive activation of Gα13 and the RhoA pathway, leading to increased SRE (serum-response element)-dependent transcription; the zinc-dependent immediate activation of GPR39 involves Gq-mediated increases in cytosolic Ca2+ and Gs coupling leading to increased cAMP levels. We used the cytosolic and soluble C-terminus of GPR39 in a Y2H (yeast-2-hybrid) screen for interacting proteins, thus identifying PKIB (protein kinase A inhibitor β). Co-expression of GPR39 with PKIB increased the protective activity of GPR39 via the constitutive, but not the ligand-mediated, pathway. PKIB inhibits protein kinase A by direct interaction with its pseudosubstrate domain; mutation of this domain abolished the inhibitory activity of PKIB on protein kinase A activity, but had no effect on the interaction with GPR39, cell protection and induction of SRE-dependent transcription. Zinc caused dissociation of PKIB from GPR39, thereby liberating it to associate with protein kinase A and inhibit its activity, which would result in a negative-feedback loop with the ability to limit activation of the Gs pathway by zinc.
Collapse
|
50
|
Cohen L, Sekler I, Hershfinkel M. The zinc sensing receptor, ZnR/GPR39, controls proliferation and differentiation of colonocytes and thereby tight junction formation in the colon. Cell Death Dis 2014; 5:e1307. [PMID: 24967969 PMCID: PMC4611734 DOI: 10.1038/cddis.2014.262] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/11/2014] [Accepted: 05/05/2014] [Indexed: 12/21/2022]
Abstract
The intestinal epithelium is a renewable tissue that requires precise balance between proliferation and differentiation, an essential process for the formation of a tightly sealed barrier. Zinc deficiency impairs the integrity of the intestinal epithelial barrier and is associated with ulcerative and diarrheal pathologies, but the mechanisms underlying the role of Zn2+ are not well understood. Here, we determined a role of the colonocytic Zn2+ sensing receptor, ZnR/GPR39, in mediating Zn2+-dependent signaling and regulating the proliferation and differentiation of colonocytes. Silencing of ZnR/GPR39 expression attenuated Zn2+-dependent activation of ERK1/2 and AKT as well as downstream activation of mTOR/p70S6K, pathways that are linked with proliferation. Consistently, ZnR/GPR39 silencing inhibited HT29 and Caco-2 colonocyte proliferation, while not inducing caspase-3 cleavage. Remarkably, in differentiating HT29 colonocytes, silencing of ZnR/GPR39 expression inhibited alkaline phosphatase activity, a marker of differentiation. Furthermore, Caco-2 colonocytes showed elevated expression of ZnR/GPR39 during differentiation, whereas silencing of ZnR/GPR39 decreased monolayer transepithelial electrical resistance, suggesting compromised barrier formation. Indeed, silencing of ZnR/GPR39 or chelation of Zn2+ by the cell impermeable chelator CaEDTA was followed by impaired expression of the junctional proteins, that is, occludin, zonula-1 (ZO-1) and E-cadherin. Importantly, colon tissues of GPR39 knockout mice also showed a decrease in expression levels of ZO-1 and occludin compared with wildtype mice. Altogether, our results indicate that ZnR/GPR39 has a dual role in promoting proliferation of colonocytes and in controlling their differentiation. The latter is followed by ZnR/GPR39-dependent expression of tight junctional proteins, thereby leading to formation of a sealed intestinal epithelial barrier. Thus, ZnR/GPR39 may be a therapeutic target for promoting epithelial function and tight junction barrier integrity during ulcerative colon diseases.
Collapse
Affiliation(s)
- L Cohen
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - I Sekler
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - M Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|