1
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
2
|
Merzeban DH, El Amin Ali AM, Hammad RO, Elmahdi MH, Sofi MA, Mahmoud RH, Metwally SM, El Ebiary AM. Differential effects of liraglutide naltrexone/bupropion, and caloric restriction on metabolic parameters and beta-cell regeneration in type 2 diabetic rat model: role of beta arrestin 1. J Mol Histol 2024; 56:50. [PMID: 39704859 DOI: 10.1007/s10735-024-10326-x] [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: 09/24/2024] [Accepted: 11/30/2024] [Indexed: 12/21/2024]
Abstract
Traditional antidiabetic treatments often carry the risk of beta-cell exhaustion, highlighting the need for therapies that promote beta-cell regeneration. This study investigates the comparative effects of Liraglutide, naltrexone/bupropion (NTX + BUP), and caloric restriction on metabolic control and beta-cell regeneration in a rat model of obese type 2 diabetes. Fifty male albino rats were randomized into five groups: normal control, diabetic control, diabetic + caloric restriction (50%), diabetic + NTX + BUP (4 mg/45 mg /kg/day orally), and diabetic + liraglutide (0.3 mg/kg/day, S.C). Body weight, BMI, serum glucose, insulin, lipid profile, atherogenic indices, beta-arrestin-1 levels, pancreatic histopathology, and immunohistochemical staining for insulin and Ki67 were assessed. All interventions significantly improved body weight, BMI, glycemic control, lipid profiles (except HDL), and atherogenic indices compared to the diabetic control group. NTX + BUP and caloric restriction resulted in greater weight loss compared to liraglutide. Of note, liraglutide significantly decreased β-arrestin-1 levels compared to both NTX + BUP and caloric restriction. Furthermore, liraglutide and caloric restriction significantly increased anti-insulin antibodies and Ki67 indicating beta-cell regeneration, while NTX + BUP showed insignificant effects. Thus we can conclude that, while NTX + BUP demonstrates efficacy in improving metabolic parameters in obese type 2 diabetic rats, it shows limitations in promoting beta-cell regeneration compared to liraglutide and caloric restriction.
Collapse
Affiliation(s)
- Dina H Merzeban
- The Departments of Medical Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt.
| | - Amani M El Amin Ali
- The Departments of Medical Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Reem O Hammad
- The Departments of Medical Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Mohamed H Elmahdi
- The Departments of Anatomical Pathology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa A Sofi
- The Departments of Histology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Rania H Mahmoud
- The Departments of Biochemistry and Molecular Biology, Fayoum University, Fayoum, Egypt
| | - Sayed M Metwally
- The Departments of Pharmacology and Toxicology, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Ahmed M El Ebiary
- The Departments of Medical Physiology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
3
|
Dai P, Chen C, Yu J, Ma C, Zhang X. New insights into sperm physiology regulation: Enlightenment from G-protein-coupled receptors. Andrology 2024; 12:1253-1271. [PMID: 38225815 DOI: 10.1111/andr.13593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/26/2023] [Accepted: 12/29/2023] [Indexed: 01/17/2024]
Abstract
BACKGROUND G-protein-coupled receptors are critical in many physiological and pathological processes in various organs. Serving as the control panel for sensing extracellular stimuli, G-protein-coupled receptors recognise various ligands, including light, temperature, odours, pheromones, hormones, neurotransmitters, chemokines, etc. Most recently, G-protein-coupled receptors residing in spermatozoa have been found to be indispensable for sperm function. OBJECTIVE Here, we have summarised cutting-edge findings on the functional mechanisms of G-protein-coupled receptors that are known to be associated with sperm functions and the activation of their downstream effectors, providing new insights into the roles of G-protein-coupled receptors in sperm physiology. RESULTS Emerging studies hint that alterations in G-protein-coupled receptors could affect sperm function, implicating their role in fertility, but solid evidence needs to be continuing excavated with various means. Several members of the G-protein-coupled receptor superfamily, including olfactory receptors, opsins, orphan G-protein-coupled receptors, CXC chemokine receptor 4, CC chemokine receptor 5 and CC chemokine receptor 6 as well as their downstream effector β-arrestins, etc., were suggested to be essential for sperm motility, capacitation, thermotaxis, chemotaxis, Ca2+ influx through CatSper channel and fertilisation capacity. CONCLUSION The present review provides a comprehensive overview of studies describing G-protein-coupled receptors and their potential action in sperm function. We also present a critical discussion of these issues, and a possible framework for future investigations on the diverse ligands, biological functions and cell signalling of G-protein-coupled receptors in spermatozoa. Here, the G-protein-coupled receptors and their related G proteins that specifically were identified in spermatozoa were summarised, and provided references valuable for further illumination, despite the evidence that is not overwhelming in most cases.
Collapse
Affiliation(s)
- Pengyuan Dai
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Chen Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Jingyan Yu
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Chaoye Ma
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| | - Xiaoning Zhang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, PR China
| |
Collapse
|
4
|
Zhao S, Jia N, Shen Z, Pei C, Huang D, Liu J, Wang Y, Shi S, Wang X, Wang M, He Y, Wang Z. Pretreatment with Notoginsenoside R1 attenuates high-altitude hypoxia-induced cardiac injury via activation of the ERK1/2-P90RSK-Bad signaling pathway in rats. Phytother Res 2023; 37:4522-4539. [PMID: 37313866 DOI: 10.1002/ptr.7923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/19/2023] [Accepted: 05/30/2023] [Indexed: 06/15/2023]
Abstract
High-altitude cardiac injury (HACI) is one of the common tissue injuries caused by high-altitude hypoxia that may be life threatening. Notoginsenoside R1 (NG-R1), a major saponin of Panax notoginseng, exerts anti-oxidative, anti-inflammatory, and anti-apoptosis effects, protecting the myocardium from hypoxic injury. This study aimed to investigate the protective effect and molecular mechanism of NG-R1 against HACI. We simulated a 6000 m environment for 48 h in a hypobaric chamber to create a HACI rat model. Rats were pretreated with NG-R1 (50, 100 mg/kg) or dexamethasone (4 mg/kg) for 3 days and then placed in the chamber for 48 h. The effect of NG-R1 was evaluated by changes in Electrocardiogram parameters, histopathology, cardiac biomarkers, oxidative stress and inflammatory indicators, key protein expression, and immunofluorescence. U0126 was used to verify whether the anti-apoptotic effect of NG-R1 was related to the activation of ERK pathway. Pretreatment with NG-R1 can improve abnormal cardiac electrical conduction and alleviate high-altitude-induced tachycardia. Similar to dexamethasone, NG-R1 can improve pathological damage, reduce the levels of cardiac injury biomarkers, oxidative stress, and inflammatory indicators, and down-regulate the expression of hypoxia-related proteins HIF-1α and VEGF. In addition, NG-R1 reduced cardiomyocyte apoptosis by down-regulating the expression of apoptotic proteins Bax, cleaved caspase 3, cleaved caspase 9, and cleaved PARP1 and up-regulating the expression of anti-apoptotic protein Bcl-2 through activating the ERK1/2-P90RSK-Bad pathway. In conclusion, NG-R1 prevented HACI and suppressed apoptosis via activation of the ERK1/2-P90RSK-Bad pathway, indicating that NG-R1 has therapeutic potential to treat HACI.
Collapse
Affiliation(s)
- Sijing Zhao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- School of Traditional Chinese Medicine, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Nan Jia
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zherui Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Caixia Pei
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Demei Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Junling Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yilan Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shihua Shi
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaomin Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Mingjie Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yacong He
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhenxing Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Liang N, Sun S, Li Z, Wu T, Zhang C, Xin T. CCKAR is a biomarker for prognosis and asynchronous brain metastasis of non-small cell lung cancer. Front Oncol 2023; 12:1098728. [PMID: 36733361 PMCID: PMC9886659 DOI: 10.3389/fonc.2022.1098728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Background Non-small cell lung cancer (NSCLC) is the most common histological type of lung cancer, and brain metastasis (BM) is the most lethal complication of NSCLC. The predictive biomarkers and risk factors of asynchronous BM are still unknown. Materials and methods A total of 203 patients with NSCLC were enrolled into our cohort and followed up. The clinicopathological factors such as tumor size, T stage, lymphatic invasion, metastasis and asynchronous BM were investigated. CCKAR expression in NSCLC and resected BM was assessed by IHC, and CCKAR mRNAs in NSCLC and para-tumor tissues were estimated by qRT-PCR. The correlations between CCKAR expression, BM and other clinicopathological factors were assessed by chi-square test, and prognostic significance of CCKAR was estimated by univariate and multivariate analyses. Results CCKAR was highly expressed in NSCLC tissues compared with para-tumor tissues. CCKAR expression in NSCLC was significantly associated with asynchronous BM. The BM percentages for NSCLC patients with low and high CCKAR were surprisingly 5.2% and 66.6%, respectively. CCKAR expression and BM were unfavorable factors predicting unfavorable outcome of NSCLC. Moreover, CCKAR expression in NSCLC was an independent risk factor of asynchronous BM. Conclusions CCKAR is a prognostic biomarker of NSCLC. CCKAR expression in NSCLC is positively associated with asynchronous BM, and is a risk factor of asynchronous BM from NSCLC.
Collapse
Affiliation(s)
- Nan Liang
- Department of Neurosurgery, the Second Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Suohui Sun
- Department of Neurosurgery, the Second Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Zheng Li
- Department of Neurosurgery, the Second Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tao Wu
- Department of Neurosurgery, the Second Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Chunpu Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Tao Xin
- Department of Neurosurgery, the First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China,*Correspondence: Tao Xin, ,
| |
Collapse
|
6
|
Jong YI, Harmon SK, O'Malley KL. GPCR
Signaling from Intracellular Membranes. GPCRS AS THERAPEUTIC TARGETS 2022:216-298. [DOI: 10.1002/9781119564782.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
7
|
Du YQ, Sha XY, Cheng J, Wang J, Lin JY, An WT, Pan W, Zhang LJ, Tao XN, Xu YF, Jia YL, Yang Z, Xiao P, Liu M, Sun JP, Yu X. Endogenous Lipid-GPR120 Signaling Modulates Pancreatic Islet Homeostasis to Different Extents. Diabetes 2022; 71:1454-1471. [PMID: 35472681 DOI: 10.2337/db21-0794] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022]
Abstract
Long-chain fatty acids (LCFAs) are not only energy sources but also serve as signaling molecules. GPR120, an LCFA receptor, plays key roles in maintaining metabolic homeostasis. However, whether endogenous ligand-GPR120 circuits exist and how such circuits function in pancreatic islets are unclear. Here, we found that endogenous GPR120 activity in pancreatic δ-cells modulated islet functions. At least two unsaturated LCFAs, oleic acid (OA) and linoleic acid (LA), were identified as GPR120 agonists within pancreatic islets. These two LCFAs promoted insulin secretion by inhibiting somatostatin secretion and showed bias activation of GPR120 in a model system. Compared with OA, LA exerted higher potency in promoting insulin secretion, which is dependent on β-arrestin2 function. Moreover, GPR120 signaling was impaired in the diabetic db/db model, and replenishing OA and LA improved islet function in both the db/db and streptozotocin-treated diabetic models. Consistently, the administration of LA improved glucose metabolism in db/db mice. Collectively, our results reveal that endogenous LCFA-GPR120 circuits exist and modulate homeostasis in pancreatic islets. The contributions of phenotype differences caused by different LCFA-GPR120 circuits within islets highlight the roles of fine-tuned ligand-receptor signaling networks in maintaining islet homeostasis.
Collapse
Affiliation(s)
- Ya-Qin Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xue-Ying Sha
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Jie Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wen-Tao An
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Wei Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Li-Jun Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao-Na Tao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Yun-Fei Xu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Ying-Li Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhao Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Peng Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
8
|
Structural insights into human brain-gut peptide cholecystokinin receptors. Cell Discov 2022; 8:55. [PMID: 35672283 PMCID: PMC9174195 DOI: 10.1038/s41421-022-00420-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 05/11/2022] [Indexed: 11/26/2022] Open
Abstract
The intestinal hormone and neuromodulator cholecystokinin (CCK) receptors CCK1R and CCK2R act as a signaling hub in brain–gut axis, mediating digestion, emotion, and memory regulation. CCK receptors exhibit distinct preferences for ligands in different posttranslational modification (PTM) states. CCK1R couples to Gs and Gq, whereas CCK2R primarily couples to Gq. Here we report the cryo-electron microscopy (cryo-EM) structures of CCK1R–Gs signaling complexes liganded either by sulfated cholecystokinin octapeptide (CCK-8) or a CCK1R-selective small-molecule SR146131, and CCK2R–Gq complexes stabilized by either sulfated CCK-8 or a CCK2R-selective ligand gastrin-17. Our structures reveal a location-conserved yet charge-distinct pocket discriminating the effects of ligand PTM states on receptor subtype preference, the unique pocket topology underlying selectivity of SR146131 and gastrin-17, the conformational changes in receptor activation, and key residues contributing to G protein subtype specificity, providing multiple structural templates for drug design targeting the brain–gut axis.
Collapse
|
9
|
Pan X, Tao S, Tong N. Potential Therapeutic Targeting Neurotransmitter Receptors in Diabetes. Front Endocrinol (Lausanne) 2022; 13:884549. [PMID: 35669692 PMCID: PMC9163348 DOI: 10.3389/fendo.2022.884549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/19/2022] [Indexed: 12/04/2022] Open
Abstract
Neurotransmitters are signaling molecules secreted by neurons to coordinate communication and proper function among different sections in the central neural system (CNS) by binding with different receptors. Some neurotransmitters as well as their receptors are found in pancreatic islets and are involved in the regulation of glucose homeostasis. Neurotransmitters can act with their receptors in pancreatic islets to stimulate or inhibit the secretion of insulin (β cell), glucagon (α cell) or somatostatin (δ cell). Neurotransmitter receptors are either G-protein coupled receptors or ligand-gated channels, their effects on blood glucose are mainly decided by the number and location of them in islets. Dysfunction of neurotransmitters receptors in islets is involved in the development of β cell dysfunction and type 2 diabetes (T2D).Therapies targeting different transmitter systems have great potential in the prevention and treatment of T2D and other metabolic diseases.
Collapse
Affiliation(s)
- Xiaohui Pan
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| | - Shibing Tao
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Department of Endocrinology, Ziyang First People’s Hospital, Ziyang, China
| | - Nanwei Tong
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Diabetes and Islet Transplantation, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
10
|
Kim HT, Desouza AH, Umhoefer H, Han J, Anzia L, Sacotte SJ, Williams RA, Blumer JT, Bartosiak JT, Fontaine DA, Baan M, Kibbe CR, Davis DB. Cholecystokinin attenuates β-cell apoptosis in both mouse and human islets. Transl Res 2022; 243:1-13. [PMID: 34740874 PMCID: PMC9504967 DOI: 10.1016/j.trsl.2021.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/23/2021] [Accepted: 10/24/2021] [Indexed: 11/17/2022]
Abstract
Loss of functional pancreatic β-cell mass and increased β-cell apoptosis are fundamental to the pathophysiology of type 1 and type 2 diabetes. Pancreatic islet transplantation has the potential to cure type 1 diabetes but is often ineffective due to the death of the islet graft within the first few years after transplant. Therapeutic strategies to directly target pancreatic β-cell survival are needed to prevent and treat diabetes and to improve islet transplant outcomes. Reducing β-cell apoptosis is also a therapeutic strategy for type 2 diabetes. Cholecystokinin (CCK) is a peptide hormone typically produced in the gut after food intake, with positive effects on obesity and glucose metabolism in mouse models and human subjects. We have previously shown that pancreatic islets also produce CCK. The production of CCK within the islet promotes β-cell survival in rodent models of diabetes and aging. We demonstrate a direct effect of CCK to reduce cytokine-mediated apoptosis in a β-cell line and in isolated mouse islets in a receptor-dependent manner. However, whether CCK can protect human β-cells was previously unknown. Here, we report that CCK can also reduce cytokine-mediated apoptosis in isolated human islets and CCK treatment in vivo decreases β-cell apoptosis in human islets transplanted into the kidney capsule of diabetic NOD/SCID mice. Collectively, these data identify CCK as a novel therapy that can directly promote β-cell survival in human islets and has therapeutic potential to preserve β-cell mass in diabetes and as an adjunct therapy after transplant.
Collapse
Affiliation(s)
- Hung Tae Kim
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Arnaldo H Desouza
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heidi Umhoefer
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jeeyoung Han
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Lucille Anzia
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Steven J Sacotte
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rashaun A Williams
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Joseph T Blumer
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jacob T Bartosiak
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Danielle A Fontaine
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mieke Baan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carly R Kibbe
- Department of Human Biology, University of Wisconsin-Green Bay, Green Bay, Wisconsin.
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, University of Wisconsin-Madison, Madison, Wisconsin; William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin.
| |
Collapse
|
11
|
Insects as a New Complex Model in Hormonal Basis of Obesity. Int J Mol Sci 2021; 22:ijms222011066. [PMID: 34681728 PMCID: PMC8540125 DOI: 10.3390/ijms222011066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 11/30/2022] Open
Abstract
Nowadays, one of the biggest problems in healthcare is an obesity epidemic. Consumption of cheap and low-quality energy-rich diets, low physical activity, and sedentary work favor an increase in the number of obesity cases within many populations/nations. This is a burden on society, public health, and the economy with many deleterious consequences. Thus, studies concerning this disorder are extremely needed, including searching for new, effective, and fitting models. Obesity may be related, among other factors, to disrupting adipocytes activity, disturbance of metabolic homeostasis, dysregulation of hormonal balance, cardiovascular problems, or disorders in nutrition which may lead to death. Because of the high complexity of obesity, it is not easy to find an ideal model for its studies which will be suitable for genetic and physiological analysis including specification of different compounds’ (hormones, neuropeptides) functions, as well as for signaling pathways analysis. In recent times, in search of new models for human diseases there has been more and more attention paid to insects, especially in neuro-endocrine regulation. It seems that this group of animals might also be a new model for human obesity. There are many arguments that insects are a good, multidirectional, and complex model for this disease. For example, insect models can have similar conservative signaling pathways (e.g., JAK-STAT signaling pathway), the presence of similar hormonal axis (e.g., brain–gut axis), or occurrence of structural and functional homologues between neuropeptides (e.g., neuropeptide F and human neuropeptide Y, insulin-like peptides, and human insulin) compared to humans. Here we give a hint to use insects as a model for obesity that can be used in multiple ways: as a source of genetic and peptidomic data about etiology and development correlated with obesity occurrence as well as a model for novel hormonal-based drug activity and their impact on mechanism of disease occurrence.
Collapse
|
12
|
Lin JY, Yang Z, Yang C, Du JX, Yang F, Cheng J, Pan W, Zhang SJ, Yan X, Wang J, Wang J, Tie L, Yu X, Chen X, Sun JP. An ionic lock and a hydrophobic zipper mediate the coupling between an insect pheromone receptor BmOR3 and downstream effectors. J Biol Chem 2021; 297:101160. [PMID: 34480896 PMCID: PMC8477192 DOI: 10.1016/j.jbc.2021.101160] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 01/14/2023] Open
Abstract
Pheromone receptors (PRs) recognize specific pheromone compounds to guide the behavioral outputs of insects, which are the most diverse group of animals on earth. The activation of PRs is known to couple to the calcium permeability of their coreceptor (Orco) or putatively with G proteins; however, the underlying mechanisms of this process are not yet fully understood. Moreover, whether this transverse seven transmembrane domain (7TM)-containing receptor is able to couple to arrestin, a common effector for many conventional 7TM receptors, is unknown. Herein, using the PR BmOR3 from the silk moth Bombyx mori and its coreceptor BmOrco as a template, we revealed that an agonist-induced conformational change of BmOR3 was transmitted to BmOrco through transmembrane segment 7 from both receptors, resulting in the activation of BmOrco. Key interactions, including an ionic lock and a hydrophobic zipper, are essential in mediating the functional coupling between BmOR3 and BmOrco. BmOR3 also selectively coupled with Gi proteins, which was dispensable for BmOrco coupling. Moreover, we demonstrated that trans-7TM BmOR3 recruited arrestin in an agonist-dependent manner, which indicates an important role for BmOR3–BmOrco complex formation in ionotropic functions. Collectively, our study identified the coupling of G protein and arrestin to a prototype trans-7TM PR, BmOR3, and provided important mechanistic insights into the coupling of active PRs to their downstream effectors, including coreceptors, G proteins, and arrestin.
Collapse
Affiliation(s)
- Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Chan Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ji-Xiang Du
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jie Cheng
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wei Pan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shi-Jie Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xu Yan
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jia Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Jin Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China.
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, China.
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
13
|
Tanday N, English A, Lafferty RA, Flatt PR, Irwin N. Benefits of Sustained Upregulated Unimolecular GLP-1 and CCK Receptor Signalling in Obesity-Diabetes. Front Endocrinol (Lausanne) 2021; 12:674704. [PMID: 34054734 PMCID: PMC8160446 DOI: 10.3389/fendo.2021.674704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Combined activation of GLP-1 and CCK1 receptors has potential to synergistically augment the appetite-suppressive and glucose homeostatic actions of the individual parent peptides. In the current study, pancreatic beta-cell benefits of combined GLP-1 and CCK1 receptor upregulation were established, before characterising bioactivity and antidiabetic efficacy of an acylated dual-acting GLP-1/CCK hybrid peptide, namely [Lys12Pal]Ex-4/CCK. Both exendin-4 and CCK exhibited (p<0.001) proliferative and anti-apoptotic effects in BRIN BD11 beta-cells. Proliferative benefits were significantly (p<0.01) augmented by combined peptide treatment when compared to either parent peptide alone. These effects were linked to increases (p<0.001) in GLUT2 and glucokinase beta-cell gene expression, with decreased (p<0.05-p<0.001) expression of NFκB and BAX. [Lys12Pal]Ex-4/CCK exhibited prominent insulinotropic actions in vitro, coupled with beneficial (p<0.001) satiety and glucose homeostatic effects in the mice, with bioactivity evident 24 h after administration. Following twice daily injection of [Lys12Pal]Ex-4/CCK for 28 days in diabetic high fat fed (HFF) mice with streptozotocin (STZ)-induced compromised beta-cells, there were clear reductions (p<0.05-p<0.001) in energy intake and body weight. Circulating glucose was returned to lean control concentrations, with associated increases (p<0.001) in plasma and pancreatic insulin levels. Glucose tolerance and insulin secretory responsiveness were significantly (p<0.05-p<0.001) improved by hybrid peptide therapy. In keeping with this, evaluation of pancreatic histology revealed restoration of normal islet alpha- to beta-cell ratios and reduction (p<0.01) in centralised islet glucagon staining. Improvements in pancreatic islet morphology were associated with increased (p<0.05) proliferation and reduced (p<0.001) apoptosis of beta-cells. Together, these data highlight the effectiveness of sustained dual GLP-1 and CCK1 receptor activation by [Lys12Pal]Ex-4/CCK for the treatment of obesity-related diabetes.
Collapse
MESH Headings
- Animals
- Biomarkers/blood
- Blood Glucose/analysis
- Body Weight
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat
- Exenatide/pharmacology
- Glucagon-Like Peptide 1/genetics
- Glucagon-Like Peptide 1/metabolism
- Hypoglycemic Agents/pharmacology
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Obesity/physiopathology
- Peptide Fragments/pharmacology
- Receptors, Cholecystokinin/genetics
- Receptors, Cholecystokinin/metabolism
- Up-Regulation
Collapse
|
14
|
Neurochemical regulators of food behavior for pharmacological treatment of obesity: current status and future prospects. Future Med Chem 2020; 12:1865-1884. [PMID: 33040605 DOI: 10.4155/fmc-2019-0361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In recent decades, obesity has become a pandemic disease and appears to be an ultimate medical and social problem. Existing antiobesity drugs show low efficiency and a wide variety of side effects. In this review, we discuss possible mechanisms underlying brain-gut-adipose tissue axis, as well as molecular biochemical characteristics of various neurochemical regulators of body weight and appetite. Multiple brain regions are responsible for eating behavior, hedonic eating and food addiction. The existing pharmacological targets for treatment of obesity were reviewed as well.
Collapse
|
15
|
Liu Q, He QT, Lyu X, Yang F, Zhu ZL, Xiao P, Yang Z, Zhang F, Yang ZY, Wang XY, Sun P, Wang QW, Qu CX, Gong Z, Lin JY, Xu Z, Song SL, Huang SM, Guo SC, Han MJ, Zhu KK, Chen X, Kahsai AW, Xiao KH, Kong W, Li FH, Ruan K, Li ZJ, Yu X, Niu XG, Jin CW, Wang J, Sun JP. DeSiphering receptor core-induced and ligand-dependent conformational changes in arrestin via genetic encoded trimethylsilyl 1H-NMR probe. Nat Commun 2020; 11:4857. [PMID: 32978402 PMCID: PMC7519161 DOI: 10.1038/s41467-020-18433-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 08/12/2020] [Indexed: 01/11/2023] Open
Abstract
Characterization of the dynamic conformational changes in membrane protein signaling complexes by nuclear magnetic resonance (NMR) spectroscopy remains challenging. Here we report the site-specific incorporation of 4-trimethylsilyl phenylalanine (TMSiPhe) into proteins, through genetic code expansion. Crystallographic analysis revealed structural changes that reshaped the TMSiPhe-specific amino-acyl tRNA synthetase active site to selectively accommodate the trimethylsilyl (TMSi) group. The unique up-field 1H-NMR chemical shift and the highly efficient incorporation of TMSiPhe enabled the characterization of multiple conformational states of a phospho-β2 adrenergic receptor/β-arrestin-1(β-arr1) membrane protein signaling complex, using only 5 μM protein and 20 min of spectrum accumulation time. We further showed that extracellular ligands induced conformational changes located in the polar core or ERK interaction site of β-arr1 via direct receptor transmembrane core interactions. These observations provided direct delineation and key mechanism insights that multiple receptor ligands were able to induce distinct functionally relevant conformational changes of arrestin.
Collapse
Affiliation(s)
- Qi Liu
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Qing-Tao He
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xiaoxuan Lyu
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Zhong-Liang Zhu
- School of Life Sciences, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Feng Zhang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Zhao-Ya Yang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Xiao-Yan Wang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Peng Sun
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, 30 Xiaohongshan Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Qian-Wen Wang
- Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, 30 Xiaohongshan Road, Wuchang District, Wuhan, Hubei, 430071, China
| | - Chang-Xiu Qu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Zheng Gong
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Jing-Yu Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Zhen Xu
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Shao-le Song
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Shen-Ming Huang
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Sheng-Chao Guo
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Ming-Jie Han
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 Xiqi Road, Airport Economic Zone, Dongli District, Tianjin, 300308, China
| | - Kong-Kai Zhu
- School of Biological Science and Technology, University of Jinan, 336 Nanxinzhuangxi Road, Shizhong District, Jinan, 250022, China
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Alem W Kahsai
- Duke University, School of Medicine, Durham, NC, 27705, USA
| | - Kun-Hong Xiao
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Wei Kong
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Fa-Hui Li
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China
| | - Ke Ruan
- Hefei National Laboratory for Physical Science at the Microscale, University of Science and Technology of China, 443 Huangshan Road, Hefei, Anhui, 230027, China
| | - Zi-Jian Li
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Xiao-Gang Niu
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, School of Life Sciences, Peking University, Beijing, 100084, China
| | - Chang-Wen Jin
- Beijing Nuclear Magnetic Resonance Center, College of Chemistry and Molecular Engineering, School of Life Sciences, Peking University, Beijing, 100084, China
| | - Jiangyun Wang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang district, Beijing, 100101, China.
- College of Life Sciences and School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo college of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, 15 Xueyuan Road, Haidian District, Beijing, 100191, China.
| |
Collapse
|
16
|
Wang C, Yu H, Wei L, Zhang J, Hong M, Chen L, Dong X, Fu L. Protective effect of cholecystokinin octapeptide on angiotensin II-induced apoptosis in H9c2 cardiomyoblast cells. J Cell Biochem 2020; 121:3560-3569. [PMID: 31886572 DOI: 10.1002/jcb.29639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
Cholecystokinin (CCK) and its receptors are expressed in mammalian cardiomyocytes and are involved in cardiovascular system regulation; however, the exact effect and underlying mechanism of CCK in cardiomyocyte apoptosis remain to be elucidated. We examined whether sulfated CCK octapeptide (CCK-8) protects H9c2 cardiomyoblast cells against angiotensin II (Ang II)-induced apoptosis. The H9c2 cardiomyoblasts were subjected to Ang II with or without CCK-8 and the viability and apoptotic rate were detected using a Cell Counting Kit-8 assay, Hoechst 33342 staining, terminal deoxyribonucleotide transferase-mediated nick-end labeling assays, and flow cytometry. In addition, specific antiapoptotic mechanisms of CCK-8 were investigated using specific CCK1 (Devazepide) or CCK2 (L365260) receptor antagonists, or the PI3K inhibitor LY294002. The expression of CCK, CCK1 receptor, CCK2 receptor, Akt, p-Akt, Bad, p-Bad, Bax, Bcl-2, and caspase-3 were detected by Western blot analysis and real-time polymerase chain reaction. We found that CCK and its receptor messenger RNA (mRNA) and protein are expressed in H9c2 cardiomyoblasts. Ang II-induced increased levels of CCK mRNA and protein expression and decreased levels of CCK1 receptor protein and mRNA. Pretreatment of CCK-8 attenuated Ang II-induced cell toxicity and apoptosis. In addition, pretreatment of H9c2 cells with CCK-8 markedly induced expression of p-Akt, p-bad, and Bcl-2 and decreased the expression levels of Bax and caspase-3. The protective effects of CCK-8 were partly abolished by Devazepide or LY294002. Our results suggest that CCK-8 protects H9c2 cardiomyoblasts from Ang II-induced apoptosis partly via activation of the CCK1 receptor and the phosphatidyqinositol-3 kinase/protein kinase B (PI3K/Akt) signaling pathway.
Collapse
Affiliation(s)
- Can Wang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huan Yu
- Department of Anesthesiology, The Fifth Hospital of Harbin City, Harbin, China
| | - Limu Wei
- Department of Internal Medicine, The First People's Hospital of Nanning, Nanning, China
| | - Jingqi Zhang
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingyang Hong
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lin Chen
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoying Dong
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lu Fu
- Laboratory of Cardiovascular Internal Medicine Department, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
17
|
Cell active and functionally-relevant small-molecule agonists of calcitonin receptor. Bioorg Chem 2020; 96:103596. [PMID: 32004895 DOI: 10.1016/j.bioorg.2020.103596] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/18/2020] [Accepted: 01/19/2020] [Indexed: 12/15/2022]
Abstract
The natural calcitonin (CT) receptor and its peptide agonists are considered validated targets for drug discovery. A small molecule agonist, SUN-B8155, has previously been shown to efficiently activate cellular CTR. Herein, we report the synthesis of a series of compounds (S8155 1-9) derived from SUN-B8155, and investigate the structural-functional relationship, bias properties and their cellular activity profile. We discover that the N-hydroxyl group from the pyridone ring is required for G protein activity and its affinity to the CT receptor. Among the compounds studied, S8155-7 exhibits improved G protein activity while S8155-4 displays a significant β-arrestin-2 signaling bias. Finally, we show that both S8155-4 and S8155-7 inhibit tumour cell invasion through CTR activation. These two compounds are anticipated to find extensive applications in chemical biology research as well drug development efforts targeting CT receptor.
Collapse
|
18
|
Hwang HJ, Yang YR, Kim HY, Choi Y, Park KS, Lee H, Ma JS, Yamamoto M, Kim J, Chae YC, Choi JH, Cocco L, Berggren PO, Jang HJ, Suh PG. Phospholipase C‐β1 potentiates glucose‐stimulated insulin secretion. FASEB J 2019; 33:10668-10679. [DOI: 10.1096/fj.201802732rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Hyeon-Jeong Hwang
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Yong Ryoul Yang
- Aging Research CenterKorea Research Institute of Bioscience and BiotechnologyDaejeonSouth Korea
| | - Hye Yun Kim
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Yoonji Choi
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Kyoung-Su Park
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Ho Lee
- Cancer Experimental Resources BranchNational Cancer CenterGoyang-siSouth Korea
| | - Ji Su Ma
- Department of ImmunoparasitologyResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Masahiro Yamamoto
- Department of ImmunoparasitologyResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Jaeyoon Kim
- Department of Molecular Medicine and SurgeryThe Rolf Luft Research Center for Diabetes and EndocrinologyKarolinska InstitutetStockholmSweden
- Division of Integrative Biosciences and BiotechnologyPohang University of Science and TechnologyPohangSouth Korea
| | - Young Chan Chae
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Jang Hyun Choi
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
- Korea Mouse Phenotyping CenterUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Lucio Cocco
- Department of Biomedical SciencesSignalling LaboratoryUniversity of BolognaBolognaItaly
| | - Per-Olof Berggren
- Department of Molecular Medicine and SurgeryThe Rolf Luft Research Center for Diabetes and EndocrinologyKarolinska InstitutetStockholmSweden
- Division of Integrative Biosciences and BiotechnologyPohang University of Science and TechnologyPohangSouth Korea
| | - Hyun-Jun Jang
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| | - Pann-Ghill Suh
- School of Life SciencesUlsan National Institute of Science and TechnologyUlsanSouth Korea
| |
Collapse
|
19
|
Ahmed Abdalhamid Osman M, Sun YJ, Li RJ, Lin H, Zeng DM, Chen XY, He D, Feng HW, Yang Z, Wang J, Wu C, Cui M, Sun JP, Huo Y, Yu X. Deletion of pancreatic β-cell adenosine kinase improves glucose homeostasis in young mice and ameliorates streptozotocin-induced hyperglycaemia. J Cell Mol Med 2019; 23:4653-4665. [PMID: 31044530 PMCID: PMC6584724 DOI: 10.1111/jcmm.14216] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 01/14/2019] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Severe reduction in the β‐cell number (collectively known as the β‐cell mass) contributes to the development of both type 1 and type 2 diabetes. Recent pharmacological studies have suggested that increased pancreatic β‐cell proliferation could be due to specific inhibition of adenosine kinase (ADK). However, genetic evidence for the function of pancreatic β‐cell ADK under physiological conditions or in a pathological context is still lacking. In this study, we crossed mice carrying LoxP‐flanked Adk gene with Ins2‐Cre mice to acquire pancreatic β ‐cell ADK deficiency (Ins2‐Cre±Adkfl/fl) mice. Our results revealed that Ins2‐Cre+/‐Adkfl/fl mice showed improved glucose metabolism and β‐cell mass in younger mice, but showed normal activity in adult mice. Moreover, Ins2‐Cre±Adkfl/fl mice were more resistant to streptozotocin (STZ) induced hyperglycaemia and pancreatic β‐cell damage in adult mice. In conclusion, we found that ADK negatively regulates β‐cell replication in young mice as well as under pathological conditions, such as STZ induced pancreatic β‐cell damage. Our study provided genetic evidence that specific inhibition of pancreatic β‐cell ADK has potential for anti‐diabetic therapy.
Collapse
Affiliation(s)
- Makawi Ahmed Abdalhamid Osman
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China.,Department of Physiology, Faculty of Medicine and Health Sciences, University of Dongola, Dongola, Sudan
| | - Yu-Jing Sun
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China.,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Rui-Jia Li
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China.,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Hui Lin
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Dong-Mei Zeng
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Xin-Yu Chen
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Dongfang He
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China.,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Hui-Wei Feng
- The Second Hospital of Shangdong University, Jinan, Shandong, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Jin Wang
- Department of Pharmacology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Min Cui
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China.,Department of Biochemistry, School of Medicine, Duke University, Durham, North Carolina
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Xiao Yu
- Department of Physiology and Pathophysiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
20
|
Li N, Yang Z, Li Q, Yu Z, Chen X, Li JC, Li B, Ning SL, Cui M, Sun JP, Yu X. Ablation of somatostatin cells leads to impaired pancreatic islet function and neonatal death in rodents. Cell Death Dis 2018; 9:682. [PMID: 29880854 PMCID: PMC5992210 DOI: 10.1038/s41419-018-0741-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 02/08/2023]
Abstract
The somatostatin (SST)-secreting cells were mainly distributed in the pancreatic islets, brain, stomach and intestine in mammals and have many physiological functions. In particular, the SST-secreting δ cell is the third most common cell type in the islets of Langerhans. Recent studies have suggested that dysregulation of paracrine interaction between the pancreatic δ cells and β cells results in impaired glucose homeostasis and contributes to diabetes development. However, direct evidence of the functional importance of SST cells in glucose homeostasis control is still lacking. In the present study, we specifically ablated SST-secreting cells by crossing Sst-cre transgenic mice with R26 DTA mice (Sst Cre R26 DTA ). The Sst Cre R26 DTA mice exhibited neonatal death. The life spans of these mice with severe hypoglycemia were extended by glucose supplementation. Moreover, we observed that SST cells deficiency led to increased insulin content and excessive insulin release, which might contribute to the observed hypoglycemia. Unexpectedly, although SST is critical for the regulation of insulin content, factors other than SST that are produced by pancreatic δ cells via their endogenous corticotropin-releasing hormone receptor 2 (CRHR2) activity play the main roles in maintaining normal insulin release, as well as neonatal glucose homeostasis in the resting state. Taken together, our results identified that the SST cells in neonatal mouse played critical role in control of insulin release and normal islet function. Moreover, we provided direct in vivo evidence of the functional importance of the SST cells, which are essential for neonatal survival and the maintenance of glucose homeostasis.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Zhao Yang
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Qing Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Zhen Yu
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Xu Chen
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Jia-Cheng Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Bo Li
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Shang-Lei Ning
- Department of General Surgery, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong, 250012, China
| | - Min Cui
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China
| | - Jin-Peng Sun
- Department of Biochemistry and Molecular Biology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China.,School of Medicine, Duke University, Durham, North Carolina, 27705, USA
| | - Xiao Yu
- Key Laboratory of Experimental Teratology of the Ministry of Education and Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, Shandong, 250012, China.
| |
Collapse
|
21
|
Gupta MK, Mohan ML, Naga Prasad SV. G Protein-Coupled Receptor Resensitization Paradigms. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 339:63-91. [PMID: 29776605 DOI: 10.1016/bs.ircmb.2018.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cellular responses to extracellular milieu/environment are driven by cell surface receptors that transmit the signal into the cells resulting in a synchronized and measured response. The ability to provide such exquisite responses to changes in external environment is mediated by the tight and yet, deliberate regulation of cell surface receptor function. In this regard, the seven transmembrane G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors that regulate responses like cardiac contractility, vision, and olfaction including platelet activation. GPCRs regulate these plethora of events through GPCR-activation, -desensitization, and -resensitization. External stimuli (ligands or agonists) activate GPCR initiating downstream signals. The activated GPCR undergoes inactivation or desensitization by phosphorylation and binding of β-arrestin resulting in diminution of downstream signals. The desensitized GPCRs are internalized into endosomes, wherein they undergo dephosphorylation or resensitization by protein phosphatase to be recycled back to the cell membrane as naïve GPCR ready for the next wave of stimuli. Despite the knowledge that activation, desensitization, and resensitization shoulder an equal role in maintaining GPCR function, major advances have been made in understanding activation and desensitization compared to resensitization. However, increasing evidence shows that resensitization is exquisitely regulated process, thereby contributing to the dynamic regulation of GPCR function. In recognition of these observations, in this chapter we discuss the key advances on the mechanistic underpinning that drive and regulate GPCR function with a focus on resensitization.
Collapse
Affiliation(s)
- Manveen K Gupta
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Maradumane L Mohan
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Sathyamangla V Naga Prasad
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|
22
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
23
|
Zhang DL, Sun YJ, Ma ML, Wang YJ, Lin H, Li RR, Liang ZL, Gao Y, Yang Z, He DF, Lin A, Mo H, Lu YJ, Li MJ, Kong W, Chung KY, Yi F, Li JY, Qin YY, Li J, Thomsen ARB, Kahsai AW, Chen ZJ, Xu ZG, Liu M, Li D, Yu X, Sun JP. Gq activity- and β-arrestin-1 scaffolding-mediated ADGRG2/CFTR coupling are required for male fertility. eLife 2018; 7:e33432. [PMID: 29393851 PMCID: PMC5839696 DOI: 10.7554/elife.33432] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/30/2018] [Indexed: 12/23/2022] Open
Abstract
Luminal fluid reabsorption plays a fundamental role in male fertility. We demonstrated that the ubiquitous GPCR signaling proteins Gq and β-arrestin-1 are essential for fluid reabsorption because they mediate coupling between an orphan receptor ADGRG2 (GPR64) and the ion channel CFTR. A reduction in protein level or deficiency of ADGRG2, Gq or β-arrestin-1 in a mouse model led to an imbalance in pH homeostasis in the efferent ductules due to decreased constitutive CFTR currents. Efferent ductule dysfunction was rescued by the specific activation of another GPCR, AGTR2. Further mechanistic analysis revealed that β-arrestin-1 acts as a scaffold for ADGRG2/CFTR complex formation in apical membranes, whereas specific residues of ADGRG2 confer coupling specificity for different G protein subtypes, this specificity is critical for male fertility. Therefore, manipulation of the signaling components of the ADGRG2-Gq/β-arrestin-1/CFTR complex by small molecules may be an effective therapeutic strategy for male infertility.
Collapse
Affiliation(s)
- Dao-Lai Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yu-Jing Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Ming-Liang Ma
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yi-jing Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Hui Lin
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Rui-Rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Zong-Lai Liang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yuan Gao
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Dong-Fang He
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Amy Lin
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Hui Mo
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yu-Jing Lu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Meng-Jing Li
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Wei Kong
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and PathophysiologySchool of Basic Medical Sciences, Peking UniversityBeijingChina
| | | | - Fan Yi
- Department of PharmacologyShandong University School of MedicineJinanChina
| | - Jian-Yuan Li
- Key Laboratory of Male Reproductive Health, National Research Institute for Family PlanningNational Health and Family Planning CommissionBeijingChina
| | - Ying-Ying Qin
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanChina
| | - Jingxin Li
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Alex R B Thomsen
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Alem W Kahsai
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Zi-Jiang Chen
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanChina
| | - Zhi-Gang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental BiologyShandong University School of Life SciencesJinanChina
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesInstitute of Biomedical Sciences, East China Normal UniversityShanghaiChina
- Department of Molecular and Cellular Medicine, Institute of Biosciences and TechnologyTexas A&M University Health Science CenterHoustonUnited States
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesInstitute of Biomedical Sciences, East China Normal UniversityShanghaiChina
| | - Xiao Yu
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| |
Collapse
|
24
|
Abstract
OBJECTIVES Modulation of cholecystokinin (CCK) receptors has been shown to influence pancreatic endocrine function. METHODS We assessed the impact of the CCKA and CCKB receptor modulators, (pGlu-Gln)-CCK-8 and gastrin-17, respectively, on β-cell secretory function, proliferation and apoptosis and glucose tolerance, and investigating alterations of CCK and gastrin islet expression in diabetes. RESULTS Initially, the presence of CCK and gastrin, and expression of their receptors were evidenced in β-cell lines and mouse islets. (pGlu-Gln)-CCK-8 and gastrin-17 stimulated insulin secretion from BRIN-BD11 and 1.1B4 β-cells, associated with no effect on membrane potential or [Ca]i. Only (pGlu-Gln)-CCK-8 possessed insulin secretory actions in isolated islets. In agreement, (pGlu-Gln)-CCK-8 improved glucose disposal and glucose-induced insulin release in mice. In addition, (pGlu-Gln)-CCK-8 evoked clear satiety effects. Interestingly, islet colocalization of CCK with glucagon was elevated in streptozotocin- and hydrocortisone-induced diabetic mice, whereas gastrin coexpression in α cells was reduced. In contrast, gastrin colocalization within β-cells was higher in diabetic mice, while CCK coexpression with insulin was decreased in insulin-deficient mice. (pGlu-Gln)-CCK-8 and gastrin-17 also augmented human and rodent β-cell proliferation and offered protection against streptozotocin-induced β-cell cytotoxicity. CONCLUSIONS We highlight the direct involvement of CCKA and CCKB receptors in pancreatic β-cell function and survival.
Collapse
|
25
|
Munakata Y, Yamada T, Imai J, Takahashi K, Tsukita S, Shirai Y, Kodama S, Asai Y, Sugisawa T, Chiba Y, Kaneko K, Uno K, Sawada S, Hatakeyama H, Kanzaki M, Miyazaki JI, Oka Y, Katagiri H. Olfactory receptors are expressed in pancreatic β-cells and promote glucose-stimulated insulin secretion. Sci Rep 2018; 8:1499. [PMID: 29367680 PMCID: PMC5784078 DOI: 10.1038/s41598-018-19765-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/04/2018] [Indexed: 11/25/2022] Open
Abstract
Olfactory receptors (ORs) mediate olfactory chemo-sensation in OR neurons. Herein, we have demonstrated that the OR chemo-sensing machinery functions in pancreatic β-cells and modulates insulin secretion. First, we found several OR isoforms, including OLFR15 and OLFR821, to be expressed in pancreatic islets and a β-cell line, MIN6. Immunostaining revealed OLFR15 and OLFR821 to be uniformly expressed in pancreatic β-cells. In addition, mRNAs of Olfr15 and Olfr821 were detected in single MIN6 cells. These results indicate that multiple ORs are simultaneously expressed in individual β-cells. Octanoic acid, which is a medium-chain fatty acid contained in food and reportedly interacts with OLFR15, potentiated glucose-stimulated insulin secretion (GSIS), thereby improving glucose tolerance in vivo. GSIS potentiation by octanoic acid was confirmed in isolated pancreatic islets and MIN6 cells and was blocked by OLFR15 knockdown. While Gαolf expression was not detectable in β-cells, experiments using inhibitors and siRNA revealed that the pathway dependent on phospholipase C-inositol triphosphate, rather than cAMP-protein kinase A, mediates GSIS potentiation via OLFR15. These findings suggest that the OR system in pancreatic β-cells has a chemo-sensor function allowing recognition of environmental substances obtained from food, and potentiates insulin secretion in a cell-autonomous manner, thereby modulating systemic glucose metabolism.
Collapse
Affiliation(s)
- Yuichiro Munakata
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Tetsuya Yamada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan. .,Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.
| | - Junta Imai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Kei Takahashi
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Sohei Tsukita
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yuta Shirai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Shinjiro Kodama
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yoichiro Asai
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Takashi Sugisawa
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yumiko Chiba
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Keizo Kaneko
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Kenji Uno
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Shojiro Sawada
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hiroyasu Hatakeyama
- Graduate School of Biomedical Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Makoto Kanzaki
- Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.,Graduate School of Biomedical Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Jun-Ichi Miyazaki
- Division of Stem Cell Regulation Research, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
| | - Yoshitomo Oka
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Hideki Katagiri
- Department of Metabolism and Diabetes, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.,Center for Metabolic Diseases, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan.,Japan Agency for Medical Research and Development (AMED), CREST, Chiyoda-ku, Tokyo, 100-0004, Japan
| |
Collapse
|
26
|
Niu S, Li H, Chen W, Zhao J, Gao L, Bo T. Beta-Arrestin 1 Mediates Liver Thyrotropin Regulation of Cholesterol Conversion Metabolism via the Akt-Dependent Pathway. Int J Endocrinol 2018; 2018:4371396. [PMID: 29853881 PMCID: PMC5954953 DOI: 10.1155/2018/4371396] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/18/2018] [Accepted: 03/31/2018] [Indexed: 11/18/2022] Open
Abstract
After activation, G protein-coupled receptors (GPCRs) are desensitized by β-arrestins (ARRBs). Moreover, ARRBs can initiate a second wave of signaling independent of G proteins. Thyroid-stimulating hormone receptor (TSHR) is one of the GPCR members. In our previous study, TSHR was identified in the liver; the major role of TSHR in cholesterol metabolism was illustrated, as TSH could regulate hepatic cholesterol metabolism via cAMP/PKA/CREB/HMGCR and SREBP2/HNF4α/CYP7A1 pathways. It has been reported that ARRB2 predominates over ARRB1 in TSHR internalization. However, the significance of ARRBs in TSH-initiated cholesterol metabolism has not been illustrated. In our study, the effects of ARRBs on TSH-regulated cholesterol metabolism are investigated. ARRB1/2 was genetically inactivated in C57BL/6 mice and HepG2 cell line, respectively. Cholesterol levels in arrestin-knockout mice and arrestin-knockdown cells were measured. Molecules participating in cholesterol metabolism were analyzed. It turned out that deficiencies in ARRB1 led to decreased cholesterol levels and decreased TSH-stimulated AKT phosphorylation. Subsequently, the inhibitory effect on CYP7A1 by SREBP2 was reduced due to lowered mature SREBP2 level. Other than the failures of TSH in ARRB-knockdown cells, the AKT activator SC79 could enhance AKT phosphorylation and mature SREBP2 level. Our results demonstrate that ARRBs, especially ARRB1, are involved in TSH-regulated cholesterol metabolism through the AKT pathway.
Collapse
Affiliation(s)
- Shaona Niu
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
- Department of Endocrinology, Lin Yi People's Hospital Affiliated to Shandong University, Linyi, Shandong 276003, China
| | - Hui Li
- Medical College, Shandong University, Jinan, Shandong 250012, China
| | - Wenbin Chen
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Jiajun Zhao
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
- Department of Endocrinology and Metabolism, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Ling Gao
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| | - Tao Bo
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Institute of Endocrinology and Metabolism, Shandong Academy of Clinical Medicine, Jinan, Shandong 250021, China
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, China
| |
Collapse
|
27
|
Li Z. A Chinese Perspective on Receptors and Receptor Regulation. Mol Pharmacol 2017; 92:185-187. [PMID: 28765267 DOI: 10.1124/mol.117.109587] [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: 06/05/2017] [Accepted: 06/06/2017] [Indexed: 11/22/2022] Open
Abstract
A receptor is a protein molecule that receives chemical signals from outside a cell, which enables the cell to respond to the signal molecule. Receptors mediate numerous important physiologic effects upon binding extracellular agonists. However, sustained activation of the receptor may lead to pathologic effects. Cells can regulate the number and function of receptors to alter their sensitivity to different molecules by a feedback mechanism, such as change in the receptor conformation, uncoupling of the receptor effector molecules, receptor sequestration, etc. In this special issue, some Chinese scientists were invited to contribute impactful discoveries and insightful reviews in the field of molecular pharmacology, especially receptor and receptor regulation.
Collapse
Affiliation(s)
- Zijian Li
- Institute of Vascular Medicine, Cardiology Department, Peking University Third Hospital, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Ministry of Health, Beijing Key Laboratory of Cardiovascular Receptors Research and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| |
Collapse
|
28
|
Yang Z, Yang F, Zhang D, Liu Z, Lin A, Liu C, Xiao P, Yu X, Sun JP. Phosphorylation of G Protein-Coupled Receptors: From the Barcode Hypothesis to the Flute Model. Mol Pharmacol 2017; 92:201-210. [PMID: 28246190 DOI: 10.1124/mol.116.107839] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/23/2017] [Indexed: 02/14/2025] Open
Abstract
Seven transmembrane G protein-coupled receptors (GPCRs) are often phosphorylated at the C terminus and on intracellular loops in response to various extracellular stimuli. Phosphorylation of GPCRs by GPCR kinases and certain other kinases can promote the recruitment of arrestin molecules. The arrestins critically regulate GPCR functions not only by mediating receptor desensitization and internalization, but also by redirecting signaling to G protein-independent pathways via interactions with numerous downstream effector molecules. Accumulating evidence over the past decade has given rise to the phospho-barcode hypothesis, which states that ligand-specific phosphorylation patterns of a receptor direct its distinct functional outcomes. Our recent work using unnatural amino acid incorporation and fluorine-19 nuclear magnetic resonance (19F-NMR) spectroscopy led to the flute model, which provides preliminary insight into the receptor phospho-coding mechanism, by which receptor phosphorylation patterns are recognized by an array of phosphate-binding pockets on arrestin and are translated into distinct conformations. These selective conformations are recognized by various effector molecules downstream of arrestin. The phospho-barcoding mechanism enables arrestin to recognize a wide range of phosphorylation patterns of GPCRs, contributing to their diverse functions.
Collapse
Affiliation(s)
- Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Daolai Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Zhixin Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Amy Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Chuan Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| |
Collapse
|
29
|
Reiter E, Ayoub MA, Pellissier LP, Landomiel F, Musnier A, Tréfier A, Gandia J, De Pascali F, Tahir S, Yvinec R, Bruneau G, Poupon A, Crépieux P. β-arrestin signalling and bias in hormone-responsive GPCRs. Mol Cell Endocrinol 2017; 449:28-41. [PMID: 28174117 DOI: 10.1016/j.mce.2017.01.052] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) play crucial roles in the ability of target organs to respond to hormonal cues. GPCRs' activation mechanisms have long been considered as a two-state process connecting the agonist-bound receptor to heterotrimeric G proteins. This view is now challenged as mounting evidence point to GPCRs being connected to large arrays of transduction mechanisms involving heterotrimeric G proteins as well as other players. Amongst the G protein-independent transduction mechanisms, those elicited by β-arrestins upon their recruitment to the active receptors are by far the best characterized and apply to most GPCRs. These concepts, in conjunction with remarkable advances made in the field of GPCR structural biology and biophysics, have supported the notion of ligand-selective signalling also known as pharmacological bias. Interestingly, recent reports have opened intriguing prospects to the way β-arrestins control GPCR-mediated signalling in space and time within the cells. In the present paper, we review the existing evidence linking endocrine-related GPCRs to β-arrestin recruitement, signalling, pathophysiological implications and selective activation by biased ligands and/or receptor modifications. Emerging concepts surrounding β-arrestin-mediated transduction are discussed in the light of the peculiarities of endocrine systems.
Collapse
Affiliation(s)
- Eric Reiter
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France.
| | - Mohammed Akli Ayoub
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France; LE STUDIUM(®) Loire Valley Institute for Advanced Studies, 45000, Orléans, France; Biology Department, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | | | - Flavie Landomiel
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Astrid Musnier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Aurélie Tréfier
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Jorge Gandia
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | | | - Shifa Tahir
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Romain Yvinec
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Gilles Bruneau
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Anne Poupon
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Pascale Crépieux
- PRC, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| |
Collapse
|
30
|
Li Q, Cui M, Yang F, Li N, Jiang B, Yu Z, Zhang D, Wang Y, Zhu X, Hu H, Li PS, Ning SL, Wang S, Qi H, Song H, He D, Lin A, Zhang J, Liu F, Zhao J, Gao L, Yi F, Xue T, Sun JP, Gong Y, Yu X. A cullin 4B-RING E3 ligase complex fine-tunes pancreatic δ cell paracrine interactions. J Clin Invest 2017; 127:2631-2646. [PMID: 28604389 DOI: 10.1172/jci91348] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 04/20/2017] [Indexed: 12/24/2022] Open
Abstract
Somatostatin secreted by pancreatic δ cells mediates important paracrine interactions in Langerhans islets, including maintenance of glucose metabolism through the control of reciprocal insulin and glucagon secretion. Disruption of this circuit contributes to the development of diabetes. However, the precise mechanisms that control somatostatin secretion from islets remain elusive. Here, we found that a super-complex comprising the cullin 4B-RING E3 ligase (CRL4B) and polycomb repressive complex 2 (PRC2) epigenetically regulates somatostatin secretion in islets. Constitutive ablation of CUL4B, the core component of the CRL4B-PRC2 complex, in δ cells impaired glucose tolerance and decreased insulin secretion through enhanced somatostatin release. Moreover, mechanistic studies showed that the CRL4B-PRC2 complex, under the control of the δ cell-specific transcription factor hematopoietically expressed homeobox (HHEX), determines the levels of intracellular calcium and cAMP through histone posttranslational modifications, thereby altering expression of the Cav1.2 calcium channel and adenylyl cyclase 6 (AC6) and modulating somatostatin secretion. In response to high glucose levels or urocortin 3 (UCN3) stimulation, increased expression of cullin 4B (CUL4B) and the PRC2 subunit histone-lysine N-methyltransferase EZH2 and reciprocal decreases in Cav1.2 and AC6 expression were found to regulate somatostatin secretion. Our results reveal an epigenetic regulatory mechanism of δ cell paracrine interactions in which CRL4B-PRC2 complexes, Cav1.2, and AC6 expression fine-tune somatostatin secretion and facilitate glucose homeostasis in pancreatic islets.
Collapse
Affiliation(s)
- Qing Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Min Cui
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Na Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Baichun Jiang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Zhen Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Daolai Zhang
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Yijing Wang
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Xibin Zhu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Huili Hu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Pei-Shan Li
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Shang-Lei Ning
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Si Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Haibo Qi
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Hechen Song
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| | - Dongfang He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology.,Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China
| | - Amy Lin
- Duke University, School of Medicine, Durham, North Carolina, USA
| | - Jingjing Zhang
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Feng Liu
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital affiliated to Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Tian Xue
- Hefei National Laboratory for Physical Science at Microscale, School of Life Science, University of Science and Technology of China, Hefei, Anhui, China
| | - Jin-Peng Sun
- Department of Biochemistry, Shandong University School of Medicine, Jinan, Shandong, China.,Duke University, School of Medicine, Durham, North Carolina, USA
| | - Yaoqin Gong
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Genetics, and
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology
| |
Collapse
|
31
|
Marchetti P, Bugliani M, De Tata V, Suleiman M, Marselli L. Pancreatic Beta Cell Identity in Humans and the Role of Type 2 Diabetes. Front Cell Dev Biol 2017; 5:55. [PMID: 28589121 PMCID: PMC5440564 DOI: 10.3389/fcell.2017.00055] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/05/2017] [Indexed: 12/13/2022] Open
Abstract
Pancreatic beta cells uniquely synthetize, store, and release insulin. Specific molecular, functional as well as ultrastructural traits characterize their insulin secretion properties and survival phentoype. In this review we focus on human islet/beta cells, and describe the changes that occur in type 2 diabetes and could play roles in the disease as well as represent possible targets for therapeutical interventions. These include transcription factors, molecules involved in glucose metabolism and insulin granule handling. Quantitative and qualitative insulin release patterns and their changes in type 2 diabetes are also associated with ultrastructural features involving the insulin granules, the mitochondria, and the endoplasmic reticulum.
Collapse
Affiliation(s)
- Piero Marchetti
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Vincenzo De Tata
- Department of Translational Medicine, University of PisaPisa, Italy
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
32
|
Liu CH, Gong Z, Liang ZL, Liu ZX, Yang F, Sun YJ, Ma ML, Wang YJ, Ji CR, Wang YH, Wang MJ, Cui FA, Lin A, Zheng WS, He DF, Qu CX, Xiao P, Liu CY, Thomsen ARB, Joseph Cahill T, Kahsai AW, Yi F, Xiao KH, Xue T, Zhou Z, Yu X, Sun JP. Arrestin-biased AT1R agonism induces acute catecholamine secretion through TRPC3 coupling. Nat Commun 2017; 8:14335. [PMID: 28181498 PMCID: PMC5309860 DOI: 10.1038/ncomms14335] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 12/19/2016] [Indexed: 12/22/2022] Open
Abstract
Acute hormone secretion triggered by G protein-coupled receptor (GPCR) activation underlies many fundamental physiological processes. GPCR signalling is negatively regulated by β-arrestins, adaptor molecules that also activate different intracellular signalling pathways. Here we reveal that TRV120027, a β-arrestin-1-biased agonist of the angiotensin II receptor type 1 (AT1R), stimulates acute catecholamine secretion through coupling with the transient receptor potential cation channel subfamily C 3 (TRPC3). We show that TRV120027 promotes the recruitment of TRPC3 or phosphoinositide-specific phospholipase C (PLCγ) to the AT1R-β-arrestin-1 signalling complex. Replacing the C-terminal region of β-arrestin-1 with its counterpart on β-arrestin-2 or using a specific TAT-P1 peptide to block the interaction between β-arrestin-1 and PLCγ abolishes TRV120027-induced TRPC3 activation. Taken together, our results show that the GPCR-arrestin complex initiates non-desensitized signalling at the plasma membrane by coupling with ion channels. This fast communication pathway might be a common mechanism of several cellular processes.
Collapse
Affiliation(s)
- Chun-Hua Liu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
- Department of Physiology, Taishan Medical University, Taian, Shandong 271000, China
| | - Zheng Gong
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Zong-Lai Liang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Zhi-Xin Liu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Fan Yang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yu-Jing Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Ming-Liang Ma
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Yi-Jing Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Chao-Ran Ji
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yu-Hong Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Mei-Jie Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Fu-Ai Cui
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Amy Lin
- Duke University, School of Medicine, Durham, North Carolina 27705, USA
| | - Wen-Shuai Zheng
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Dong-Fang He
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Chang-xiu Qu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| | - Chuan-Yong Liu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | | | | | - Alem W. Kahsai
- Duke University, School of Medicine, Durham, North Carolina 27705, USA
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Kun-Hong Xiao
- Duke University, School of Medicine, Durham, North Carolina 27705, USA
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA
| | - Tian Xue
- Hefei National Laboratory for Physical Science at Microscale, School of Life Science, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Zhuan Zhou
- Laboratory of Cellular Biophysics and Neurodegeneration, Ying-Jie Conference Center, Peking University, Beijing 100871, China
| | - Xiao Yu
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, 44 Wenhua Xi Road, Jinan, Shandong 250012, China
| |
Collapse
|
33
|
Miller LJ, Desai AJ. Metabolic Actions of the Type 1 Cholecystokinin Receptor: Its Potential as a Therapeutic Target. Trends Endocrinol Metab 2016; 27:609-619. [PMID: 27156041 PMCID: PMC4992613 DOI: 10.1016/j.tem.2016.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/31/2016] [Accepted: 04/05/2016] [Indexed: 12/13/2022]
Abstract
Cholecystokinin (CCK) regulates appetite and reduces food intake by activating the type 1 CCK receptor (CCK1R). Attempts to develop CCK1R agonists for obesity have yielded active agents that have not reached clinical practice. Here we discuss why, along with new strategies to target CCK1R more effectively. We examine signaling events and the possibility of developing agents that exhibit ligand-directed bias, to dissociate satiety activity from undesirable side effects. Potential allosteric sites of modulation are also discussed, along with desired properties of a positive allosteric modulator (PAM) without intrinsic agonist action as another strategy to treat obesity. These new types of CCK1R-active drugs could be useful as standalone agents or as part of a rational drug combination for management of obesity.
Collapse
Affiliation(s)
- Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, 85259, USA.
| | - Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, 85259, USA
| |
Collapse
|
34
|
Linnemann AK, Davis DB. Glucagon-like peptide-1 and cholecystokinin production and signaling in the pancreatic islet as an adaptive response to obesity. J Diabetes Investig 2016; 7 Suppl 1:44-9. [PMID: 27186355 PMCID: PMC4854504 DOI: 10.1111/jdi.12465] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Precise control of blood glucose is dependent on adequate β‐cell mass and function. Thus, reductions in β‐cell mass and function lead to insufficient insulin production to meet demand, and result in diabetes. Recent evidence suggests that paracrine signaling in the islet might be important in obesity, and disruption of this signaling could play a role in the pathogenesis of diabetes. For example, we recently discovered a novel islet incretin axis where glucagon‐like peptide‐1 regulates β‐cell production of another classic gut hormone, cholecystokinin. This axis is stimulated by obesity, and plays a role in enhancing β‐cell survival. In the present review, we place our observations in the wider context of the literature on incretin regulation in the islet, and discuss the potential for therapeutic targeting of these pathways.
Collapse
Affiliation(s)
- Amelia K Linnemann
- Department of Medicine Division of Endocrinology University of Wisconsin-Madison Madison Wisconsin USA
| | - Dawn Belt Davis
- Department of MedicineDivision of EndocrinologyUniversity of Wisconsin-MadisonMadisonWisconsinUSA; William S Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| |
Collapse
|
35
|
Lavine JA, Kibbe CR, Baan M, Sirinvaravong S, Umhoefer HM, Engler KA, Meske LM, Sacotte KA, Erhardt DP, Davis DB. Cholecystokinin expression in the β-cell leads to increased β-cell area in aged mice and protects from streptozotocin-induced diabetes and apoptosis. Am J Physiol Endocrinol Metab 2015; 309:E819-28. [PMID: 26394663 PMCID: PMC4652070 DOI: 10.1152/ajpendo.00159.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022]
Abstract
Cholecystokinin (CCK) is a peptide hormone produced in the gut and brain with beneficial effects on digestion, satiety, and insulin secretion. CCK is also expressed in pancreatic β-cells, but only in models of obesity and insulin resistance. Whole body deletion of CCK in obese mice leads to reduced β-cell mass expansion and increased apoptosis. We hypothesized that islet-derived CCK is important in protection from β-cell apoptosis. To determine the specific role of β-cell-derived CCK in β-cell mass dynamics, we generated a transgenic mouse that expresses CCK in the β-cell in the lean state (MIP-CCK). Although this transgene contains the human growth hormone minigene, we saw no expression of human growth hormone protein in transgenic islets. We examined the ability of MIP-CCK mice to maintain β-cell mass when subjected to apoptotic stress, with advanced age, and after streptozotocin treatment. Aged MIP-CCK mice have increased β-cell area. MIP-CCK mice are resistant to streptozotocin-induced diabetes and exhibit reduced β-cell apoptosis. Directed CCK overexpression in cultured β-cells also protects from cytokine-induced apoptosis. We have identified an important new paracrine/autocrine effect of CCK in protection of β-cells from apoptotic stress. Understanding the role of β-cell CCK adds to the emerging knowledge of classic gut peptides in intraislet signaling. CCK receptor agonists are being investigated as therapeutics for obesity and diabetes. While these agonists clearly have beneficial effects on body weight and insulin sensitivity in peripheral tissues, they may also directly protect β-cells from apoptosis.
Collapse
Affiliation(s)
- Jeremy A Lavine
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carly R Kibbe
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mieke Baan
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sirinart Sirinvaravong
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heidi M Umhoefer
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kimberly A Engler
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Louise M Meske
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaitlyn A Sacotte
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Daniel P Erhardt
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin; Geriatric Research Education and Clinical Centers, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
36
|
Ning SL, Zheng WS, Su J, Liang N, Li H, Zhang DL, Liu CH, Dong JH, Zhang ZK, Cui M, Hu QX, Chen CC, Liu CH, Wang C, Pang Q, Chen YX, Yu X, Sun JP. Different downstream signalling of CCK1 receptors regulates distinct functions of CCK in pancreatic beta cells. Br J Pharmacol 2015; 172:5050-67. [PMID: 26248680 DOI: 10.1111/bph.13271] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 07/18/2015] [Accepted: 07/26/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Cholecystokinin (CCK) is secreted by intestinal I cells and regulates important metabolic functions. In pancreatic islets, CCK controls beta cell functions primarily through CCK1 receptors, but the signalling pathways downstream of these receptors in pancreatic beta cells are not well defined. EXPERIMENTAL APPROACH Apoptosis in pancreatic beta cell apoptosis was evaluated using Hoechst-33342 staining, TUNEL assays and Annexin-V-FITC/PI staining. Insulin secretion and second messenger production were monitored using ELISAs. Protein and phospho-protein levels were determined by Western blotting. A glucose tolerance test was carried out to examine the functions of CCK-8s in streptozotocin-induced diabetic mice. KEY RESULTS The sulfated carboxy-terminal octapeptide CCK26-33 amide (CCK-8s) activated CCK1 receptors and induced accumulation of both IP3 and cAMP. Whereas Gq -PLC-IP3 signalling was required for the CCK-8s-induced insulin secretion under low-glucose conditions, Gs -PKA/Epac signalling contributed more strongly to the CCK-8s-mediated insulin secretion in high-glucose conditions. CCK-8s also promoted formation of the CCK1 receptor/β-arrestin-1 complex in pancreatic beta cells. Using β-arrestin-1 knockout mice, we demonstrated that β-arrestin-1 is a key mediator of both CCK-8s-mediated insulin secretion and of its the protective effect against apoptosis in pancreatic beta cells. The anti-apoptotic effects of β-arrestin-1 occurred through cytoplasmic late-phase ERK activation, which activates the 90-kDa ribosomal S6 kinase-phospho-Bcl-2-family protein pathway. CONCLUSIONS AND IMPLICATIONS Knowledge of different CCK1 receptor-activated downstream signalling pathways in the regulation of distinct functions of pancreatic beta cells could be used to identify biased CCK1 receptor ligands for the development of new anti-diabetic drugs.
Collapse
Affiliation(s)
- Shang-lei Ning
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Qilu Hospital, Shandong University, Jinan, Shandong, China.,Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Wen-shuai Zheng
- Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Jing Su
- Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Nan Liang
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Hui Li
- Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Dao-lai Zhang
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China
| | - Chun-hua Liu
- Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Jun-hong Dong
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Zheng-kui Zhang
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Min Cui
- Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Qiao-Xia Hu
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Chao-chao Chen
- Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Chang-hong Liu
- Shandong Provincial Qianfoshan, Shandong University, Jinan, Shandong, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qi Pang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yu-xin Chen
- Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xiao Yu
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Qilu Hospital, Shandong University, Jinan, Shandong, China.,Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Physiology, Shandong University School of Medicine, Jinan, Shandong, China
| | - Jin-peng Sun
- Department of Biochemistry and Molecular Biology and Key Laboratory Experimental Teratology of the Ministry of Education, Jinan, Shandong, China.,Shandong Provincial School Key laboratory for Protein Science of Chronic degenerative diseases, Jinan, Shandong, China.,Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China.,Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|