1
|
Jang MH, Song J. Adenosine and adenosine receptors in metabolic imbalance-related neurological issues. Biomed Pharmacother 2024; 177:116996. [PMID: 38897158 DOI: 10.1016/j.biopha.2024.116996] [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: 04/24/2024] [Revised: 06/08/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic syndromes (e.g., obesity) are characterized by insulin resistance, chronic inflammation, impaired glucose metabolism, and dyslipidemia. Recently, patients with metabolic syndromes have experienced not only metabolic problems but also neuropathological issues, including cognitive impairment. Several studies have reported blood-brain barrier (BBB) disruption and insulin resistance in the brain of patients with obesity and diabetes. Adenosine, a purine nucleoside, is known to regulate various cellular responses (e.g., the neuroinflammatory response) by binding with adenosine receptors in the central nervous system (CNS). Adenosine has four known receptors: A1R, A2AR, A2BR, and A3R. These receptors play distinct roles in various physiological and pathological processes in the brain, including endothelial cell homeostasis, insulin sensitivity, microglial activation, lipid metabolism, immune cell infiltration, and synaptic plasticity. Here, we review the recent findings on the role of adenosine receptor-mediated signaling in neuropathological issues related to metabolic imbalance. We highlight the importance of adenosine signaling in the development of therapeutic solutions for neuropathological issues in patients with metabolic syndromes.
Collapse
Affiliation(s)
- Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, United States.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Republic of Korea.
| |
Collapse
|
2
|
Nguyen ATN, Tran QL, Baltos JA, McNeill SM, Nguyen DTN, May LT. Small molecule allosteric modulation of the adenosine A 1 receptor. Front Endocrinol (Lausanne) 2023; 14:1184360. [PMID: 37435481 PMCID: PMC10331460 DOI: 10.3389/fendo.2023.1184360] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/23/2023] [Indexed: 07/13/2023] Open
Abstract
G protein-coupled receptors (GPCRs) represent the target for approximately a third of FDA-approved small molecule drugs. The adenosine A1 receptor (A1R), one of four adenosine GPCR subtypes, has important (patho)physiological roles in humans. A1R has well-established roles in the regulation of the cardiovascular and nervous systems, where it has been identified as a potential therapeutic target for a number of conditions, including cardiac ischemia-reperfusion injury, cognition, epilepsy, and neuropathic pain. A1R small molecule drugs, typically orthosteric ligands, have undergone clinical trials. To date, none have progressed into the clinic, predominantly due to dose-limiting unwanted effects. The development of A1R allosteric modulators that target a topographically distinct binding site represent a promising approach to overcome current limitations. Pharmacological parameters of allosteric ligands, including affinity, efficacy and cooperativity, can be optimized to regulate A1R activity with high subtype, spatial and temporal selectivity. This review aims to offer insights into the A1R as a potential therapeutic target and highlight recent advances in the structural understanding of A1R allosteric modulation.
Collapse
Affiliation(s)
- Anh T. N. Nguyen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Quan L. Tran
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Jo-Anne Baltos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Samantha M. McNeill
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Diep T. N. Nguyen
- Department of Information Technology, Faculty of Engineering and Technology, Vietnam National University, Hanoi, Vietnam
| | - Lauren T. May
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
3
|
Olofinsan KA, Salau VF, Erukainure OL, Islam MS. Senna petersiana (Bolle) leaf extract modulates glycemic homeostasis and improves dysregulated enzyme activities in fructose-fed streptozotocin-induced diabetic rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 303:115998. [PMID: 36471537 DOI: 10.1016/j.jep.2022.115998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Senna petersiana (Bolle) is a native South African medicinal shrub combined locally with other plant products to manage diabetes or used as a single therapy for several other ailing conditions. AIM OF THE STUDY This study evaluated the antidiabetic and antilipidemic effects of S. petersiana leaf ethanol extract and its modulatory effects on dysregulated enzyme activities in fructose-fed streptozotocin-induced diabetic rats. MATERIALS AND METHODS Six groups of 6-weeks old male Sprague Dawley rats were used in this study. Diabetes was induced in four of the groups by injecting (i.p.) 40 mg/kg of streptozotocin after a two-weeks feeding of 10% fructose via drinking water, while animals in the two normal groups were given similar volume of vehicle buffer and normal drinking water, respectively. After the confirmation of diabetes, treatment with 150 and 300 mg/kg body weight of the ethanolic leaf extract of S. petersiana proceeded for a period of 6 weeks. RESULTS Oral administration of S. petersiana leaf extract significantly lowered blood glucose, food and liquid intake, glycosylhaemoglobin in blood, liver and cardiac biomarkers, and lipid profile in serum and atherogenic index (AIP) in both the low and high-dose treated animal groups. This was accompanied by a simultaneous increase in Homeostatic Model Assessment-beta (HOMA-β) score, serum high-density lipoproteins cholesterol (HDL-c), and insulin levels. It also improved pancreatic and serum-reduced glutathione (GSH) levels, catalase, and superoxide dismutase (SOD) enzymes activities with a simultaneous reduction in malondialdehyde (MDA) and nitric oxide (NO) concentrations. Moreover, the extract modulated dysregulated α-amylase, lipase, cholinesterase, and 5' nucleotidase enzyme activities in pancreatic tissue as well as glycogen metabolism in the liver. Analysis of the phytochemicals in the S. petersiana extract showed the presence of phytol, 4a,7,7,10a-tetramethyldodecahydrobenzo[f]-chromen-3-ol, phytol acetate, solasodine glucoside, cassine, veratramine and solasodine acetate. Amongst these compounds, solasodine glucoside had the best binding energy (ΔG) with the selected diabetes-linked enzymes via molecular docking simulation. CONCLUSION Data from this study demonstrate the antidiabetic effects of S. petersiana leaf extract via the modulation of the dysregulated indices involved in type 2 diabetes and its associated complications. Although it has been shown safe in animals, further toxicological studies are required to ensure its safety for diabetes management in humans.
Collapse
Affiliation(s)
- Kolawole A Olofinsan
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Veronica F Salau
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa
| | - Ochuko L Erukainure
- Department of Pharmacology, University of the Free State, Bloemfontein, 9300, South Africa
| | - Md Shahidul Islam
- Department of Biochemistry, University of KwaZulu-Natal, Westville Campus, Durban, 4000, South Africa.
| |
Collapse
|
4
|
Granade ME, Hargett SR, Lank DS, Lemke MC, Luse MA, Isakson BE, Bochkis IM, Linden J, Harris TE. Feeding desensitizes A1 adenosine receptors in adipose through FOXO1-mediated transcriptional regulation. Mol Metab 2022; 63:101543. [PMID: 35811051 PMCID: PMC9304768 DOI: 10.1016/j.molmet.2022.101543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 12/03/2022] Open
Abstract
OBJECTIVE Adipose tissue is a critical regulator of energy balance that must rapidly shift its metabolism between fasting and feeding to maintain homeostasis. Adenosine has been characterized as an important regulator of adipocyte metabolism primarily through its actions on A1 adenosine receptors (A1R). We sought to understand the role A1R plays specifically in adipocytes during fasting and feeding to regulate glucose and lipid metabolism. METHODS We used Adora1 floxed mice with an inducible, adiponectin-Cre to generate FAdora1-/- mice, where F designates a fat-specific deletion of A1R. We used these FAdora1-/- mice along with specific agonists and antagonists of A1R to investigate changes in adenosine signaling within adipocytes between the fasted and fed state. RESULTS We found that the adipose tissue response to adenosine is not static, but changes dynamically according to nutrient conditions through the insulin-Akt-FOXO1 axis. We show that under fasted conditions, FAdora1-/- mice had impairments in the suppression of lipolysis by insulin on normal chow and impaired glucose tolerance on high-fat diet. FAdora1-/- mice also exhibited a higher lipolytic response to isoproterenol than WT controls when fasted, however this difference was lost after a 4-hour refeeding period. We demonstrate that FOXO1 binds to the A1R promoter, and refeeding leads to a rapid downregulation of A1R transcript and desensitization of adipocytes to A1R agonism. Obesity also desensitizes adipocyte A1R, and this is accompanied by a disruption of cyclical changes in A1R transcription between fasting and refeeding. CONCLUSIONS We propose that FOXO1 drives high A1R expression under fasted conditions to limit excess lipolysis during stress and augment insulin action upon feeding. Subsequent downregulation of A1R under fed conditions leads to desensitization of these receptors in adipose tissue. This regulation of A1R may facilitate reentrance into the catabolic state upon fasting.
Collapse
Affiliation(s)
- Mitchell E Granade
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Stefan R Hargett
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Daniel S Lank
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Michael C Lemke
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Melissa A Luse
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| | - Brant E Isakson
- Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville, VA, USA
| | - Irina M Bochkis
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Joel Linden
- Department of Medicine, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, VA, USA
| | - Thurl E Harris
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
5
|
Pak ES, Cha JJ, Cha DR, Kanasaki K, Ha H. Adenosine receptors as emerging therapeutic targets for diabetic kidney disease. Kidney Res Clin Pract 2022; 41:S74-S88. [PMID: 36239063 PMCID: PMC9590297 DOI: 10.23876/j.krcp.22.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 08/07/2023] Open
Abstract
Diabetic kidney disease (DKD) is now a pandemic worldwide, and novel therapeutic options are urgently required. Adenosine, an adenosine triphosphate metabolite, plays a role in kidney homeostasis through interacting with four types of adenosine receptors (ARs): A1AR, A2AAR, A2BAR, and A3AR. Increasing evidence highlights the role of adenosine and ARs in the development and progression of DKD: 1) increased adenosine in the plasma and urine of diabetics with kidney injury, 2) increased expression of each of the ARs in diabetic kidneys, 3) the protective effect of coffee, a commonly ingested nonselective AR antagonist, on DKD, and 4) the protective effect of AR modulators in experimental DKD models. We propose AR modulators as a new therapeutic option to treat DKD. Detailed mechanistic studies on the pharmacology of AR modulators will help us to develop effective first-in-class AR modulators against DKD.
Collapse
Affiliation(s)
- Eun Seon Pak
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Jin Joo Cha
- Department of Nephrology, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Dae Ryong Cha
- Department of Nephrology, Korea University Ansan Hospital, Ansan, Republic of Korea
| | - Keizo Kanasaki
- Department of Internal Medical 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
6
|
Han L, Wang G, Zhou S, Situ C, He Z, Li Y, Qiu Y, Huang Y, Xu A, Ong MTY, Wang H, Zhang J, Wu Z. Muscle satellite cells are impaired in type 2 diabetic mice by elevated extracellular adenosine. Cell Rep 2022; 39:110884. [PMID: 35649375 DOI: 10.1016/j.celrep.2022.110884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 04/12/2022] [Accepted: 05/05/2022] [Indexed: 12/25/2022] Open
Abstract
Muscle regeneration is known to be defective under diabetic conditions. However, the underlying mechanisms remain less clear. Adult quiescent muscle satellite cells (MuSCs) from leptin-receptor-deficient (i.e., db/db) diabetic mice are defective in early activation in vivo, but not in culture, suggesting the involvement of pathogenic niche factors. Elevated extracellular adenosine (eAdo) and AMP (eAMP) are detected under diabetic conditions. eAdo and eAMP potently inhibit cell cycle re-entry of quiescent MuSCs and injury-induced muscle regeneration. Mechanistically, eAdo and eAMP engage the equilibrative Ado transporters (ENTs)-Ado kinase (ADK)-AMPK signaling axis in MuSCs to inhibit the mTORC1-dependent cell growth checkpoint. eAdo and eAMP also inhibit early activation of quiescent fibroadipogenic progenitors and human MuSCs by the same mechanism. Treatment of db/db diabetic mice with an ADK inhibitor partially rescues the activation defects of MuSCs in vivo. Thus, both ADK and ENTs represent potential therapeutic targets for restoring the regenerative functions of tissue stem cells in patients with diabetes.
Collapse
Affiliation(s)
- Lifang Han
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China
| | - Gang Wang
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China
| | - Shaopu Zhou
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China
| | - Chenghao Situ
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China
| | - Zhiming He
- Department of Chemical Pathology, the Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Li Ka Shing Institute of Health Sciences, Department of Orthopaedics and Traumatology, the Chinese University of Hong Kong, Hong Kong, China
| | - Yudan Qiu
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, the City University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Michael Tim Yun Ong
- Department of Orthopaedics and Traumatology, the Chinese University of Hong Kong, the Prince of Wales Hospital, Hong Kong, China
| | - Huating Wang
- Li Ka Shing Institute of Health Sciences, Department of Orthopaedics and Traumatology, the Chinese University of Hong Kong, Hong Kong, China
| | - Jianfa Zhang
- Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Zhenguo Wu
- Division of Life Science, the State Key Laboratory on Molecular Neuroscience, the Hong Kong University of Science & Technology, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Sanni O, Terre'Blanche G. Therapeutic potentials of agonist and antagonist of adenosine receptors in type 2 diabetes. Rev Endocr Metab Disord 2021; 22:1073-1090. [PMID: 34165671 DOI: 10.1007/s11154-021-09668-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 10/21/2022]
Abstract
Type 2 diabetes has been a global health challenge over the decades and is among the leading causes of death. Several treatment approaches have been developed, but more effective and new therapies are still needed. The role of adenosine in glucose and lipid homeostasis has offered a different therapeutic approach. Adenosine mediates its physiological role through the activation of adenosine receptors. These adenosine receptors have been implicated in glucose and lipid homeostasis. The ability of agonists and antagonists of adenosine receptors to activate or inhibit the adenosine signalling cascade and thereby affecting the balance of glucose and lipid homeostasis has challenged the studies of agonists and antagonists of adenosine receptors, both preclinical and clinical, as potential anti-diabetic drugs. This review provides a background on different anti-diabetic therapeutic approaches, outlining the role of adenosine receptors in glucose and lipid homeostasis, and mechanisms underlying the action of agonists/antagonists of adenosine receptors as a therapeutic potential towards type 2 diabetes.
Collapse
Affiliation(s)
- Olakunle Sanni
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen), School of Health Sciences. North-West University (NWU), Potchefstroom, 2357, South Africa.
| | - G Terre'Blanche
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen), School of Health Sciences. North-West University (NWU), Potchefstroom, 2357, South Africa
| |
Collapse
|
8
|
Reichert KP, Castro MFV, Assmann CE, Bottari NB, Miron VV, Cardoso A, Stefanello N, Morsch VMM, Schetinger MRC. Diabetes and hypertension: Pivotal involvement of purinergic signaling. Biomed Pharmacother 2021; 137:111273. [PMID: 33524787 PMCID: PMC7846467 DOI: 10.1016/j.biopha.2021.111273] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/11/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus (DM) and hypertension are highly prevalent worldwide health problems and frequently associated with severe clinical complications, such as diabetic cardiomyopathy, nephropathy, retinopathy, neuropathy, stroke, and cardiac arrhythmia, among others. Despite all existing research results and reasonable speculations, knowledge about the role of purinergic system in individuals with DM and hypertension remains restricted. Purinergic signaling accounts for a complex network of receptors and extracellular enzymes responsible for the recognition and degradation of extracellular nucleotides and adenosine. The main components of this system that will be presented in this review are: P1 and P2 receptors and the enzymatic cascade composed by CD39 (NTPDase; with ATP and ADP as a substrate), CD73 (5'-nucleotidase; with AMP as a substrate), and adenosine deaminase (ADA; with adenosine as a substrate). The purinergic system has recently emerged as a central player in several physiopathological conditions, particularly those linked to inflammatory responses such as diabetes and hypertension. Therefore, the present review focuses on changes in both purinergic P1 and P2 receptor expression as well as the activities of CD39, CD73, and ADA in diabetes and hypertension conditions. It can be postulated that the manipulation of the purinergic axis at different levels can prevent or exacerbate the insurgency and evolution of diabetes and hypertension working as a compensatory mechanism.
Collapse
Affiliation(s)
- Karine Paula Reichert
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Milagros Fanny Vera Castro
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Charles Elias Assmann
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Nathieli Bianchin Bottari
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vanessa Valéria Miron
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Andréia Cardoso
- Academic Coordination, Medicine, Campus Chapecó, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Naiara Stefanello
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Melchiors Morsch
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Department of Biochemistry and Molecular Biology, Post-Graduation Program of Biological Sciences: Toxicological Biochemistry, CCNE, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
9
|
Caruso V, Zuccarini M, Di Iorio P, Muhammad I, Ronci M. Metabolic Changes Induced by Purinergic Signaling: Role in Food Intake. Front Pharmacol 2021; 12:655989. [PMID: 33995077 PMCID: PMC8117016 DOI: 10.3389/fphar.2021.655989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/09/2021] [Indexed: 01/30/2023] Open
Abstract
The purinergic signalling has a well-established role in the regulation of energy homeostasis, but there is growing evidence of its implication in the control of food intake. In this review, we provide an integrative view of the molecular mechanisms leading to changes in feeding behaviour within hypothalamic neurons following purinergic receptor activation. We also highlight the importance of purinergic signalling in metabolic homeostasis and the possibility of targeting its receptors for therapeutic purposes.
Collapse
Affiliation(s)
- Vanni Caruso
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia.,Institute for Research on Pain, ISAL-Foundation, Rimini, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy
| | - Patrizia Di Iorio
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, Chieti, Italy.,Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy
| | - Ishaq Muhammad
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, TAS, Australia
| | - Maurizio Ronci
- Center for Advanced Studies and Technologies (CAST), University of Chieti-Pescara, Chieti, Italy.,Department of Pharmacy, University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
10
|
Im H, Park JH, Im S, Han J, Kim K, Lee YH. Regulatory roles of G-protein coupled receptors in adipose tissue metabolism and their therapeutic potential. Arch Pharm Res 2021; 44:133-145. [PMID: 33550564 PMCID: PMC7907040 DOI: 10.1007/s12272-021-01314-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
The high incidence of obesity has increased the need to discover new therapeutic targets to combat obesity and obesity-related metabolic diseases. Obesity is defined as an abnormal accumulation of adipose tissue, which is one of the major metabolic organs that regulate energy homeostasis. However, there are currently no approved anti-obesity therapeutics that directly target adipose tissue metabolism. With recent advances in the understanding of adipose tissue biology, molecular mechanisms involved in brown adipose tissue expansion and metabolic activation have been investigated as potential therapeutic targets to increase energy expenditure. This review focuses on G-protein coupled receptors (GPCRs) as they are the most successful class of druggable targets in human diseases and have an important role in regulating adipose tissue metabolism. We summarize recent findings on the major GPCR classes that regulate thermogenesis and mitochondrial metabolism in adipose tissue. Improved understanding of GPCR signaling pathways that regulate these processes could facilitate the development of novel pharmacological approaches to treat obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Hyeonyeong Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Ji-Hyun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seowoo Im
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Juhyeong Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Kyungmin Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yun-Hee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University; Bio-MAX Institute, Seoul National University, 29-Room # 311, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
11
|
Jain S, Jacobson KA. Purinergic signaling in diabetes and metabolism. Biochem Pharmacol 2020; 187:114393. [PMID: 33359363 DOI: 10.1016/j.bcp.2020.114393] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
Purinergic signaling, a concept originally formulated by the late Geoffrey Burnstock (1929-2020), was found to modulate pathways in every physiological system. In metabolic disorders there is a role for both adenosine receptors and P2 (nucleotide) receptors, of which there are two classes, i.e. P2Y metabotropic and P2X ionotropic receptors. The individual roles of the 19 receptors encompassed by this family have been dissected - and in many cases the effects associated with specific cell types, including adipocytes, skeletal muscle, liver cells and immune cells. It is suggested that ligands selective for each of the four adenosine receptors (A1, A2A, A2B and A3), and several of the P2 subtypes (e.g. P2Y6 or P2X7 antagonists) might have therapeutic potential for treating diabetes and obesity. This is a developing story with some conflicting conclusions relevant to drug discovery, which we summarize here.
Collapse
Affiliation(s)
- Shanu Jain
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA.
| |
Collapse
|
12
|
Eisenstein A, Chitalia SV, Ravid K. Bone Marrow and Adipose Tissue Adenosine Receptors Effect on Osteogenesis and Adipogenesis. Int J Mol Sci 2020; 21:E7470. [PMID: 33050467 PMCID: PMC7589187 DOI: 10.3390/ijms21207470] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an extracellular signaling molecule that is particularly relevant in times of cellular stress, inflammation and metabolic disturbances when the levels of the purine increase. Adenosine acts on two G-protein-coupled stimulatory and on two G-protein-coupled inhibitory receptors, which have varying expression profiles in different tissues and conditions, and have different affinities for the endogenous ligand. Studies point to significant roles of adenosine and its receptors in metabolic disease and bone health, implicating the receptors as potential therapeutic targets. This review will highlight our current understanding of the dichotomous effects of adenosine and its receptors on adipogenesis versus osteogenesis within the bone marrow to maintain bone health, as well as its relationship to obesity. Therapeutic implications will also be reviewed.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shlok V. Chitalia
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| |
Collapse
|
13
|
Olaniyi KS, Sabinari IW, Olatunji LA. Oral L-glutamine restores adenosine and glutathione content in the skeletal muscle and adipose tissue of insulin-resistant pregnant rats. Nutrition 2020; 77:110789. [PMID: 32428839 DOI: 10.1016/j.nut.2020.110789] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 01/21/2020] [Accepted: 02/07/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Mishandling of lipid and glycogen has been documented as a feature of metabolic tissues in insulin resistance-related disorders. However, reports exist detailing that L-glutamine (GLN) protects non-adipose tissue against the deleterious effects of metabolic disorders. Therefore, we hypothesized that GLN would protect skeletal muscle and adipose tissue against the deleterious effects of lipid and glycogen mishandlings by increasing adenosine and glutathione levels in pregnant rats exposed to fructose (FRU)-enriched drinks. METHODS Pregnant Wistar rats weighing 150 to 180 g were randomly assigned to control, GLN, FRU, and FRU + GLN groups (six rats/group). The groups received vehicle (P.o.), glutamine (1 g/kg), FRU (10%; w/v), and FRU + GLN, respectively, for 19 d. RESULTS Data show that FRU caused insulin resistance with corresponding increased blood glucose, circulating and pancreatic insulin levels, and lipid accumulation and glycogen depletion in skeletal muscle, but glycogen accumulation and a decreased lipid profile in adipose tissue. Adenosine and glutathione content decreased, whereas adenosine deaminase, xanthine oxidase, uric acid, and malondialdehyde concentrations increased in both tissues. In addition, glucose-6-phosphate dehydrogenase activity decreased in skeletal muscle but remained unaltered in adipose tissue. However, supplementation with GLN improved perturbed lipid and glycogen with a corresponding increase in adenosine and glutathione. CONCLUSIONS The present results collectively indicate that lipid and glycogen mishandlings caused by high gestational FRU intake result in the depletion of adenosine and glutathione in skeletal muscle and adipose tissue. These findings also suggest that L-glutamine protects against skeletal muscle and adipose tissue dysmetabolism by enhancing adenosine and glutathione.
Collapse
Affiliation(s)
- Kehinde Samuel Olaniyi
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria; Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Isaiah Woru Sabinari
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence Aderemi Olatunji
- HOPE Cardiometabolic Research Team & Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.
| |
Collapse
|
14
|
Long residence time adenosine A1 receptor agonists produce sustained wash-resistant antilipolytic effect in rat adipocytes. Biochem Pharmacol 2019; 164:45-52. [DOI: 10.1016/j.bcp.2019.03.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 03/20/2019] [Indexed: 02/06/2023]
|
15
|
Blackwood SJ, Hanya E, Katz A. Effect of postexercise temperature elevation on postexercise glycogen metabolism of isolated mouse soleus muscle. J Appl Physiol (1985) 2019; 126:1103-1109. [PMID: 30730817 DOI: 10.1152/japplphysiol.01121.2018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effects of temperature elevation after intense repeated contractions on glycogen and energy metabolism as well as contractile function of isolated mouse soleus muscle (slow twitch, oxidative) were investigated. Muscles were stimulated electrically to perform repeated tetanic contractions for 10 min at 25°C, which reduced tetanic force by ~85% and glycogen by 50%. After 120-min recovery at 25°C glycogen was fully restored (~125% of basal), whereas after recovery at 35°C glycogen decreased further (~25% of basal). Glycogen synthase fractional activity averaged 31.8 ± 3.1% (baseline = 33.8 ± 3.4%) after 120-min recovery at 25°C but was increased after recovery at 35°C (63.8 ± 4.8%; P < 0.001 vs. 25°C). Phosphorylase fractional and total activities were not affected by the higher temperature. However, recovery at 35°C resulted in a significantly higher content of the phosphorylase substrate inorganic phosphate (~20%; P < 0.01 vs. 25°C). Finally, fatigue development during a subsequent bout of repeated contractions at 25°C was similar after 120-min recovery at 25°C and 35°C. These data demonstrate that after intense contractions elevated temperature inhibits glycogen accumulation, likely by increasing the availability of the phosphorylase substrate inorganic phosphate, but has no effect on fatigue development. Thus after heat exposure phosphorylase plays a significant role in glycogen accumulation, and glycogen does not limit muscle performance in isolated mouse soleus muscle after recovery from elevated temperature. NEW & NOTEWORTHY Whether elevated temperature affects glycogen biogenesis and contractile performance of isolated slow-twitch muscle is not known. Here we show that after a bout of repeated contractions in isolated mouse soleus muscle at 25°C, increasing muscle temperature during recovery to 35°C blocked glycogen accumulation compared with recovery at 25°C. Surprisingly, during a subsequent bout of repeated contractions at 25°C, the rate of fatigue was not different between groups after recovery at the two temperatures.
Collapse
Affiliation(s)
- Sarah J Blackwood
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Ester Hanya
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Abram Katz
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
16
|
Duarte JMN, Skoug C, Silva HB, Carvalho RA, Gruetter R, Cunha RA. Impact of Caffeine Consumption on Type 2 Diabetes-Induced Spatial Memory Impairment and Neurochemical Alterations in the Hippocampus. Front Neurosci 2019; 12:1015. [PMID: 30686981 PMCID: PMC6333904 DOI: 10.3389/fnins.2018.01015] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes affects the morphology and plasticity of the hippocampus, and leads to learning and memory deficits. Caffeine has been proposed to prevent memory impairment upon multiple chronic disorders with neurological involvement. We tested whether long-term caffeine consumption prevents type 2 diabetes (T2D)-induced spatial memory impairment and hippocampal alterations, including synaptic degeneration, astrogliosis, and metabolic modifications. Control Wistar rats and Goto-Kakizaki (GK) rats that develop T2D were treated with caffeine (1 g/L in drinking water) for 4 months. Spatial memory was evaluated in a Y-maze. Hippocampal metabolic profile and glucose homeostasis were investigated by 1H magnetic resonance spectroscopy. The density of neuronal, synaptic, and glial-specific markers was evaluated by Western blot analysis. GK rats displayed reduced Y-maze spontaneous alternation and a lower amplitude of hippocampal long-term potentiation when compared to controls, suggesting impaired hippocampal-dependent spatial memory. Diabetes did not impact the relation of hippocampal to plasma glucose concentrations, but altered the neurochemical profile of the hippocampus, such as increased in levels of the osmolites taurine (P < 0.001) and myo-inositol (P < 0.05). The diabetic hippocampus showed decreased density of the presynaptic proteins synaptophysin (P < 0.05) and SNAP25 (P < 0.05), suggesting synaptic degeneration, and increased GFAP (P < 0.001) and vimentin (P < 0.05) immunoreactivities that are indicative of astrogliosis. The effects of caffeine intake on hippocampal metabolism added to those of T2D, namely reducing myo-inositol levels (P < 0.001) and further increasing taurine levels (P < 0.05). Caffeine prevented T2D-induced alterations of GFAP, vimentin and SNAP25, and improved memory deficits. We conclude that caffeine consumption has beneficial effects counteracting alterations in the hippocampus of GK rats, leading to the improvement of T2D-associated memory impairment.
Collapse
Affiliation(s)
- João M N Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Cecilia Skoug
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Henrique B Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rui A Carvalho
- Department of Life Sciences, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Department of Radiology, University of Lausanne, Lausanne, Switzerland.,Department of Radiology, University of Geneva, Geneva, Switzerland
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
17
|
Simard T, Jung R, Labinaz A, Faraz MA, Ramirez FD, Di Santo P, Pitcher I, Motazedian P, Gaudet C, Rochman R, Marbach J, Boland P, Sarathy K, Alghofaili S, Russo JJ, Couture E, Beanlands RS, Hibbert B. Adenosine as a Marker and Mediator of Cardiovascular Homeostasis: A Translational Perspective. Cardiovasc Hematol Disord Drug Targets 2019; 19:109-131. [PMID: 30318008 DOI: 10.2174/1871529x18666181011103719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 08/08/2018] [Accepted: 09/25/2018] [Indexed: 06/08/2023]
Abstract
Adenosine, a purine nucleoside, is produced broadly and implicated in the homeostasis of many cells and tissues. It signals predominantly via 4 purinergic adenosine receptors (ADORs) - ADORA1, ADORA2A, ADORA2B and ADOosine signaling, both through design as specific ADOR agonists and antagonists and as offtarget effects of existing anti-platelet medications. Despite this, adenosine has yet to be firmly established as either a therapeutic or a prognostic tool in clinical medicine to date. Herein, we provide a bench-to-bedside review of adenosine biology, highlighting the key considerations for further translational development of this proRA3 in addition to non-ADOR mediated effects. Through these signaling mechanisms, adenosine exerts effects on numerous cell types crucial to maintaining vascular homeostasis, especially following vascular injury. Both in vitro and in vivo models have provided considerable insights into adenosine signaling and identified targets for therapeutic intervention. Numerous pharmacologic agents have been developed that modulate adenmising molecule.
Collapse
Affiliation(s)
- Trevor Simard
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada
| | - Richard Jung
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada
| | - Alisha Labinaz
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | | | - F Daniel Ramirez
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Pietro Di Santo
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Ian Pitcher
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Pouya Motazedian
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, ON, Canada
| | - Chantal Gaudet
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Rebecca Rochman
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Jeffrey Marbach
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Paul Boland
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Kiran Sarathy
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Saleh Alghofaili
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Juan J Russo
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Etienne Couture
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
| | - Rob S Beanlands
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada
| | - Benjamin Hibbert
- CAPITAL research group, Division of Cardiology, University of Ottawa Heart Institute, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Canada
| |
Collapse
|
18
|
Singh A, Gibert Y, Dwyer KM. The adenosine, adrenergic and opioid pathways in the regulation of insulin secretion, beta cell proliferation and regeneration. Pancreatology 2018; 18:615-623. [PMID: 29937364 DOI: 10.1016/j.pan.2018.06.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/25/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023]
Abstract
Insulin, a key hormone produced by pancreatic beta cells precisely regulates glucose metabolism in vertebrates. In type 1 diabetes, the beta cell mass is destroyed, a process triggered by a combination of environmental and genetic factors. This ultimately results in absolute insulin deficiency and dysregulated glucose metabolism resulting in a number of detrimental pathophysiological effects. The traditional focus of treating type 1 diabetes has been to control blood sugar levels through the administration of exogenous insulin. Newer approaches aim to replace the beta cell mass through pancreatic or islet transplantation. Type 2 diabetes results from a relative insulin deficiency for the prevailing insulin resistance. Treatments are generally aimed at reducing insulin resistance and/or augmenting insulin secretion and the use of insulin itself is often required. It is increasingly being recognized that the beta cell mass is dynamic and increases insulin secretion in response to beta cell mitogens and stress signals to maintain glycemia within a very narrow physiological range. This review critically discusses the role of adrenergic, adenosine and opioid pathways and their interrelationship in insulin secretion, beta cell proliferation and regeneration.
Collapse
Affiliation(s)
- Amitoj Singh
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Yann Gibert
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia
| | - Karen M Dwyer
- Deakin University, School of Medicine, Faculty of Health, 75 Pigdons Rd, Waurn Ponds, Geelong, VIC, 3216, Australia.
| |
Collapse
|
19
|
Metabolomics coupled with pathway analysis characterizes metabolic changes in response to BDE-3 induced reproductive toxicity in mice. Sci Rep 2018; 8:5423. [PMID: 29615664 PMCID: PMC5882662 DOI: 10.1038/s41598-018-23484-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 03/06/2018] [Indexed: 12/21/2022] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) may affect male reproductive function. 4-bromodiphenyl ether (BDE-3), the photodegradation products of higher brominated PBDEs, is the most fundamental mono-BDE in environment but is less studied. The purpose of this study was to investigate the reproductive toxicity induced by BDE-3 and explore the mechanism by metabolomics approach. In this study, mice were treated intragastrically with BDE-3 for consecutive six weeks at the dosages of 0.0015, 1.5, 10 and 30 mg/kg. The reproductive toxicity was evaluated by sperm analysis and histopathology examinations. UPLC-Q-TOF/MS was applied to profile the metabolites of testis tissue, urine and serum samples in the control and BDE-3 treated mice. Results showed the sperm count was dose-dependently decreased and percentage of abnormal sperms increased by the treatment of BDE-3. Histopathology examination also revealed changes in seminiferous tubules and epididymides in BDE-3 treated mice. Metabolomics analysis revealed that different BDE-3 groups showed metabolic disturbances to varying degrees. We identified 76, 38 and 31 differential metabolites in testis tissue, urine and serum respectively. Pathway analysis revealed several pathways including Tyrosine metabolism, Purine metabolism and Riboflavin metabolism, which may give a possible explanation for the toxic mechanism of BDE-3. This study indicates that UHPLC-Q-TOFMS-based metabolomics approach provided a better understanding of PBDEs-induced toxicity dynamically.
Collapse
|
20
|
Carlström M, Larsson SC. Coffee consumption and reduced risk of developing type 2 diabetes: a systematic review with meta-analysis. Nutr Rev 2018; 76:395-417. [DOI: 10.1093/nutrit/nuy014] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Susanna C Larsson
- Unit of Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Varani K, Vincenzi F, Merighi S, Gessi S, Borea PA. Biochemical and Pharmacological Role of A1 Adenosine Receptors and Their Modulation as Novel Therapeutic Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1051:193-232. [DOI: 10.1007/5584_2017_61] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
22
|
Peleli M, Carlstrom M. Adenosine signaling in diabetes mellitus and associated cardiovascular and renal complications. Mol Aspects Med 2017; 55:62-74. [DOI: 10.1016/j.mam.2016.12.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 12/18/2016] [Accepted: 12/21/2016] [Indexed: 12/21/2022]
|
23
|
Leiva A, Guzmán-Gutiérrez E, Contreras-Duarte S, Fuenzalida B, Cantin C, Carvajal L, Salsoso R, Gutiérrez J, Pardo F, Sobrevia L. Adenosine receptors: Modulators of lipid availability that are controlled by lipid levels. Mol Aspects Med 2017; 55:26-44. [DOI: 10.1016/j.mam.2017.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 01/25/2017] [Accepted: 01/25/2017] [Indexed: 12/20/2022]
|
24
|
Pardo F, Villalobos-Labra R, Chiarello DI, Salsoso R, Toledo F, Gutierrez J, Leiva A, Sobrevia L. Molecular implications of adenosine in obesity. Mol Aspects Med 2017; 55:90-101. [PMID: 28104382 DOI: 10.1016/j.mam.2017.01.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 12/30/2016] [Accepted: 01/13/2017] [Indexed: 12/31/2022]
Abstract
Adenosine has broad activities in organisms due to the existence of multiple receptors, the differential adenosine concentrations necessary to activate these receptors and the presence of proteins able to synthetize, degrade or transport this nucleoside. All adenosine receptors have been reported to be involved in glucose homeostasis, inflammation, adipogenesis, insulin resistance, and thermogenesis, indicating that adenosine could participate in the process of obesity. Since adenosine seems to be associated with several effects, it is plausible that adenosine participates in the initiation and development of obesity or may function to prevent it. Thus, the purpose of this review was to explore the involvement of adenosine in adipogenesis, insulin resistance and thermogenesis, with the aim of understanding how adenosine could be used to avoid, treat or improve the metabolic state of obesity. Treatment with specific agonists and/or antagonists of adenosine receptors could reverse the obesity state, since adenosine receptors normalizes several mechanisms involved in obesity, such as lipolysis, insulin sensitivity and thermogenesis. Furthermore, obesity is a preventable state, and the specific activation of adenosine receptors could aid in the prevention of obesity. Nevertheless, for the treatment of obesity and its consequences, more studies and therapeutic strategies in addition to adenosine are necessary.
Collapse
Affiliation(s)
- Fabián Pardo
- Metabolic Diseases Research Laboratory, Center of Research, Development and Innovation in Health - Aconcagua Valley, San Felipe Campus, School of Medicine, Faculty of Medicine, Universidad de Valparaiso, 2172972 San Felipe, Chile; Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| | - Roberto Villalobos-Labra
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Delia I Chiarello
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Rocío Salsoso
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain
| | - Fernando Toledo
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Basic Sciences, Faculty of Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile
| | - Jaime Gutierrez
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Cellular Signaling Differentiation and Regeneration Laboratory, Health Sciences Faculty, Universidad San Sebastian, Santiago, Chile
| | - Andrea Leiva
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Luis Sobrevia
- Cellular and Molecular Physiology Laboratory, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research, Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD 4029, Queensland, Australia.
| |
Collapse
|
25
|
Pharmacological targeting of adenosine receptor signaling. Mol Aspects Med 2017; 55:4-8. [PMID: 28088486 DOI: 10.1016/j.mam.2016.12.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 12/20/2022]
Abstract
Adenosine receptor signaling plays important roles in normal physiology, but is also known to modulate the development or progression of several different diseases. The design of new, efficient, and safe pharmacological approaches to target the adenosine system may have considerable therapeutic potential, but is also associated with many challenges. This review summarizes the main challenges of adenosine receptor targeted treatment including tolerance, disease stage, cell type-specific effects, caffeine intake, adenosine level assessment and receptor distribution in vivo. Moreover, we discuss several potential ways to overcome these obstacles (i.e., the use of partial agonists, indirect receptor targeting, allosteric enhancers, prodrugs, non-receptor-mediated effects, neoreceptors, conditional knockouts). It is important to address these concerns during development of new and successful therapeutic approaches targeting the adenosine system.
Collapse
|
26
|
Westermeier F, Riquelme JA, Pavez M, Garrido V, Díaz A, Verdejo HE, Castro PF, García L, Lavandero S. New Molecular Insights of Insulin in Diabetic Cardiomyopathy. Front Physiol 2016; 7:125. [PMID: 27148064 PMCID: PMC4828458 DOI: 10.3389/fphys.2016.00125] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/22/2016] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a highly prevalent disease worldwide. Cardiovascular disorders generated as a consequence of T2DM are a major cause of death related to this disease. Diabetic cardiomyopathy (DCM) is characterized by the morphological, functional and metabolic changes in the heart produced as a complication of T2DM. This cardiac disorder is characterized by constant high blood glucose and lipids levels which eventually generate oxidative stress, defective calcium handling, altered mitochondrial function, inflammation and fibrosis. In this context, insulin is of paramount importance for cardiac contractility, growth and metabolism and therefore, an impaired insulin signaling plays a critical role in the DCM development. However, the exact pathophysiological mechanisms leading to DCM are still a matter of study. Despite the numerous questions raised in the study of DCM, there have also been important findings, such as the role of micro-RNAs (miRNAs), which can not only have the potential of being important biomarkers, but also therapeutic targets. Furthermore, exosomes also arise as an interesting variable to consider, since they represent an important inter-cellular communication mechanism and therefore, they may explain many aspects of the pathophysiology of DCM and their study may lead to the development of therapeutic agents capable of improving insulin signaling. In addition, adenosine and adenosine receptors (ARs) may also play an important role in DCM. Moreover, the possible cross-talk between insulin and ARs may provide new strategies to reverse its defective signaling in the diabetic heart. This review focuses on DCM, the role of insulin in this pathology and the discussion of new molecular insights which may help to understand its underlying mechanisms and generate possible new therapeutic strategies.
Collapse
Affiliation(s)
- Francisco Westermeier
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Jaime A Riquelme
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Mario Pavez
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Valeria Garrido
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Ariel Díaz
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Hugo E Verdejo
- Faculty of Medicine, Advanced Center for Chronic Diseases, Pontifical Catholic University of ChileSantiago, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontifical Catholic University of ChileSantiago, Chile
| | - Pablo F Castro
- Faculty of Medicine, Advanced Center for Chronic Diseases, Pontifical Catholic University of ChileSantiago, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontifical Catholic University of ChileSantiago, Chile
| | - Lorena García
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of Chile Santiago, Chile
| | - Sergio Lavandero
- Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Advanced Center for Chronic Diseases, University of ChileSantiago, Chile; Department of Internal Medicine (Division of Cardiology), University of Texas Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
27
|
Katz A. Role of reactive oxygen species in regulation of glucose transport in skeletal muscle during exercise. J Physiol 2016; 594:2787-94. [PMID: 26791627 DOI: 10.1113/jp271665] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/15/2016] [Indexed: 12/28/2022] Open
Abstract
Glucose derived from extracellular sources serves as an energy source in virtually all eukaryotic cells, including skeletal muscle. Its contribution to energy turnover increases with exercise intensity up to moderately heavy workloads. However, at very high workloads, the contribution of extracellular glucose to energy turnover is negligible, despite the high rate of glucose transport. Reactive oxygen species (ROS) are involved in the stimulation of glucose transport in isolated skeletal muscle preparations during intense repeated contractions. Consistent with this observation, heavy exercise is associated with significant production of ROS. However, during more mild to moderate stimulation or exercise conditions (in vitro, in situ and in vivo) antioxidants do not affect glucose transport. It is noteworthy that the production of ROS is limited or not observed under these conditions and that the concentration of the antioxidant used was extremely low. The results to date suggest that ROS involvement in activation of glucose transport occurs primarily during intense short-term exercise and that other mechanisms are involved during mild to moderate exercise. What remains puzzling is why ROS-mediated activation of glucose transport would occur under conditions where glucose transport is highest and utilization (i.e. phosphorylation of glucose by hexokinase) is low. Possibly ROS production is involved in priming glucose transport during heavy exercise to accelerate glycogen biogenesis during the initial recovery period after exercise, as well as altering other aspects of intracellular metabolism.
Collapse
Affiliation(s)
- Abram Katz
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, 40700, Israel
| |
Collapse
|
28
|
Ventromedial Hypothalamic Lesions Down-Regulate the Expression of Adora1 Gene in Rat Pancreatic Islets. Pancreas 2016; 45:e1-2. [PMID: 26658041 DOI: 10.1097/mpa.0000000000000390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
29
|
Eisenstein A, Patterson S, Ravid K. The Many Faces of the A2b Adenosine Receptor in Cardiovascular and Metabolic Diseases. J Cell Physiol 2015; 230:2891-7. [PMID: 25975415 DOI: 10.1002/jcp.25043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 01/09/2023]
Abstract
Modulation of the low affinity adenosine receptor subtype, the A2b adenosine receptor (A2bAR), has gained interest as a therapeutic target in various pathologic areas associated with cardiovascular disease. The actions of the A2bAR are diverse and at times conflicting depending on cell and tissue type and the timing of activation or inhibition of the receptor. The A2bAR is a promising and exciting pharmacologic target, however, a thorough understanding of A2bAR action is necessary to reach the therapeutic potential of this receptor. This review will focus on the role of the A2bAR in various cardiovascular and metabolic pathologies in which the receptor is currently being studied. We will illustrate the complexities of A2bAR signaling and highlight areas of research with potential for therapeutic development.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Shenia Patterson
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Katya Ravid
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.,Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts.,Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts.,Evans Center for Interdisciplinary Biomedical Research, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
30
|
Merighi S, Borea PA, Gessi S. Adenosine receptors and diabetes: Focus on the A2B adenosine receptor subtype. Pharmacol Res 2015; 99:229-36. [DOI: 10.1016/j.phrs.2015.06.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 06/23/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022]
|
31
|
Yang T, Gao X, Sandberg M, Zollbrecht C, Zhang XM, Hezel M, Liu M, Peleli M, Lai EY, Harris RA, Persson AEG, Fredholm BB, Jansson L, Carlström M. Abrogation of adenosine A1 receptor signalling improves metabolic regulation in mice by modulating oxidative stress and inflammatory responses. Diabetologia 2015; 58:1610-20. [PMID: 25835725 DOI: 10.1007/s00125-015-3570-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/02/2015] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Adenosine is an important regulator of metabolism; however, the role of the A1 receptor during ageing and obesity is unclear. The aim of this study was to investigate the effects of A1 signalling in modulating metabolic function during ageing. METHODS Age-matched young and aged A 1 (also known as Adora1)-knockout (A1(-/-)) and wild-type (A1(+/+)) mice were used. Metabolic regulation was evaluated by body composition, and glucose and insulin tolerance tests. Isolated islets and islet arterioles were used to detect islet endocrine and vascular function. Oxidative stress and inflammation status were measured in metabolic organs and systemically. RESULTS Advanced age was associated with both reduced glucose clearance and insulin sensitivity, as well as increased visceral adipose tissue (VAT) in A1(+/+) compared with A1(-/-) mice. Islet morphology and insulin content were similar between genotypes, but relative changes in in vitro insulin release following glucose stimulation were reduced in aged A1(+/+) compared with A1(-/-) mice. Islet arteriolar responses to angiotensin II were stronger in aged A1(+/+) mice, this being associated with increased NADPH oxidase activity. Ageing resulted in multiple changes in A1(+/+) compared with A1(-/-) mice, including enhanced NADPH oxidase-derived O2(-) formation and NADPH oxidase isoform 2 (Nox2) protein expression in pancreas and VAT; elevated levels of circulating insulin, leptin and proinflammatory cytokines (TNF-α, IL-1β, IL-6 and IL-12); and accumulation of CD4(+) T cells in VAT. This was associated with impaired insulin signalling in VAT from aged A1(+/+) mice. CONCLUSIONS/INTERPRETATION These studies emphasise that A1 receptors regulate metabolism and islet endocrine and vascular functions during ageing, including via the modulation of oxidative stress and inflammatory responses, among other things.
Collapse
Affiliation(s)
- Ting Yang
- Department of Physiology and Pharmacology, Karolinska Institutet, Nanna Svartz Väg 2, SE-171 77, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Antonioli L, Blandizzi C, Csóka B, Pacher P, Haskó G. Adenosine signalling in diabetes mellitus--pathophysiology and therapeutic considerations. Nat Rev Endocrinol 2015; 11:228-41. [PMID: 25687993 DOI: 10.1038/nrendo.2015.10] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adenosine is a key extracellular signalling molecule that regulates several aspects of tissue function by activating four G-protein-coupled receptors, A1, A2A, A2B and A1 adenosine receptors. Accumulating evidence highlights a critical role for the adenosine system in the regulation of glucose homeostasis and the pathophysiology of type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Although adenosine signalling is known to affect insulin secretion, new data indicate that adenosine signalling also contributes to the regulation of β-cell homeostasis and activity by controlling the proliferation and regeneration of these cells as well as the survival of β cells in inflammatory microenvironments. Furthermore, adenosine is emerging as a major regulator of insulin responsiveness by controlling insulin signalling in adipose tissue, muscle and liver; adenosine also indirectly mediates effects on inflammatory and/or immune cells in these tissues. This Review critically discusses the role of the adenosine-adenosine receptor system in regulating both the onset and progression of T1DM and T2DM, and the potential of pharmacological manipulation of the adenosinergic system as an approach to manage T1DM, T2DM and their associated complications.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Balázs Csóka
- Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, 185 South Orange Avenue, University Heights, Newark, NJ 07103, USA
| | - Pál Pacher
- Section on Oxidative Stress Tissue Injury, Laboratories of Physiological Studies, NIH/NIAAA, 5625 Fishers Lane, Bethesda, MD 20892, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, 185 South Orange Avenue, University Heights, Newark, NJ 07103, USA
| |
Collapse
|
33
|
Yip L, Fathman CG. Type 1 diabetes in mice and men: gene expression profiling to investigate disease pathogenesis. Immunol Res 2015; 58:340-50. [PMID: 24682832 DOI: 10.1007/s12026-014-8501-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Type 1 diabetes (T1D) is a complex polygenic disease that is triggered by various environmental factors in genetically susceptible individuals. The emphasis placed on genome-wide association studies to explain the genetics of T1D has failed to advance our understanding of T1D pathogenesis or identify biomarkers of disease progression or therapeutic targets. Using the nonobese diabetic (NOD) mouse model of T1D and the non-disease prone congenic NOD.B10 mice, our laboratory demonstrated striking tissue-specific and age-dependent changes in gene expression during disease progression. We established a "roadmap" of differential gene expression and used this to identify candidate genes in mice (and human orthologs) that play a role in disease pathology. Here, we describe two genes, Deformed epidermal autoregulatory factor 1 (Deaf1) and Adenosine A1 receptor (Adora1), that are differentially expressed and alternatively spliced in the pancreatic lymph nodes or islets of NOD mice and T1D patients to form dominant-negative non-functional isoforms. Loss of Deaf1 function leads to reduced peripheral tissue antigen expression in lymph node stromal cells and may contribute to a breakdown in peripheral tolerance, while reduced Adora1 function results in an early intrinsic alpha cell defect that may explain the hyperglucagonemia and resulting beta cell stress observed prior to the onset of diabetes. Remarkably, both genes were also alternatively spliced in the same tissues of auto-antibody positive prediabetic patients, and these splicing events resulted in similar downstream effects as those seen in NOD mice. These findings demonstrate the value of gene expression profiling in studying disease pathogenesis in T1D.
Collapse
Affiliation(s)
- Linda Yip
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, 269 Campus Drive, CCSR Room 2225, Stanford, CA, 94305-5166, USA
| | | |
Collapse
|
34
|
Szkudelski T, Szkudelska K. Regulatory role of adenosine in insulin secretion from pancreatic β-cells--action via adenosine A₁ receptor and beyond. J Physiol Biochem 2014; 71:133-40. [PMID: 25432862 DOI: 10.1007/s13105-014-0371-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 11/17/2014] [Indexed: 01/04/2023]
Abstract
Under physiological conditions, insulin secretion from pancreatic β-cells is tightly regulated by different factors, including nutrients, nervous system, and other hormones. Pancreatic β-cells are also influenced by paracrine and autocrine interactions. The results of rodent studies indicate that adenosine is present within pancreatic islets and is implicated in the regulation of insulin secretion; however, effects depend on adenosine and glucose concentrations. Moreover, species differences in adenosine action were found. In rat islets, low adenosine was demonstrated to decrease glucose-induced insulin secretion and this effect is mediated via adenosine A1 receptor. In the presence of high adenosine concentrations, other mechanisms are activated and glucose-induced insulin secretion is increased. It is also well established that suppression of adenosine action increases insulin-secretory response of β-cells to glucose. In mouse islets, low adenosine concentrations do not significantly affect insulin secretion. However, in the presence of higher adenosine concentrations, potentiation of glucose-induced insulin secretion was demonstrated. It is also known that upon stimulation of insulin secretion, both rat and mouse islets release ATP. In rat islets, ATP undergoes extracellular conversion to adenosine. However, mouse islets are unable to convert extracellularly ATP to adenosine and adenosine arises from intracellular ATP degradation.
Collapse
Affiliation(s)
- Tomasz Szkudelski
- Department of Animal Physiology and Biochemistry, Poznan University of Life Sciences, Wolynska 35, 60-637, Poznan, Poland,
| | | |
Collapse
|
35
|
Sakamoto S, Miyaji T, Hiasa M, Ichikawa R, Uematsu A, Iwatsuki K, Shibata A, Uneyama H, Takayanagi R, Yamamoto A, Omote H, Nomura M, Moriyama Y. Impairment of vesicular ATP release affects glucose metabolism and increases insulin sensitivity. Sci Rep 2014; 4:6689. [PMID: 25331291 PMCID: PMC4204045 DOI: 10.1038/srep06689] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/17/2014] [Indexed: 12/04/2022] Open
Abstract
Neuroendocrine cells store ATP in secretory granules and release it along with hormones that may trigger a variety of cellular responses in a process called purinergic chemical transmission. Although the vesicular nucleotide transporter (VNUT) has been shown to be involved in vesicular storage and release of ATP, its physiological relevance in vivo is far less well understood. In Vnut knockout (Vnut(-/-)) mice, we found that the loss of functional VNUT in adrenal chromaffin granules and insulin granules in the islets of Langerhans led to several significant effects. Vesicular ATP accumulation and depolarization-dependent ATP release were absent in the chromaffin granules of Vnut(-/-) mice. Glucose-responsive ATP release was also absent in pancreatic β-cells in Vnut(-/-) mice, while glucose-responsive insulin secretion was enhanced to a greater extent than that in wild-type tissue. Vnut(-/-) mice exhibited improved glucose tolerance and low blood glucose upon fasting due to increased insulin sensitivity. These results demonstrated an essential role of VNUT in vesicular storage and release of ATP in neuroendocrine cells in vivo and suggest that vesicular ATP and/or its degradation products act as feedback regulators in catecholamine and insulin secretion, thereby regulating blood glucose homeostasis.
Collapse
Affiliation(s)
- Shohei Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Takaaki Miyaji
- Advanced Research Science Center, Okayama University, Okayama 700-8530, JAPAN
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| | - Reiko Ichikawa
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Akira Uematsu
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Ken Iwatsuki
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Atsushi Shibata
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Hisayuki Uneyama
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Ryoichi Takayanagi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Akitsugu Yamamoto
- Faculty of Bioscience, Nagahama Institute of Bio-science and Technology, Nagahama 526-0829, JAPAN
| | - Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Yoshinori Moriyama
- Advanced Research Science Center, Okayama University, Okayama 700-8530, JAPAN
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| |
Collapse
|
36
|
Burnstock G. Purinergic signalling in endocrine organs. Purinergic Signal 2014; 10:189-231. [PMID: 24265070 PMCID: PMC3944044 DOI: 10.1007/s11302-013-9396-x] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/24/2013] [Indexed: 01/08/2023] Open
Abstract
There is widespread involvement of purinergic signalling in endocrine biology. Pituitary cells express P1, P2X and P2Y receptor subtypes to mediate hormone release. Adenosine 5'-triphosphate (ATP) regulates insulin release in the pancreas and is involved in the secretion of thyroid hormones. ATP plays a major role in the synthesis, storage and release of catecholamines from the adrenal gland. In the ovary purinoceptors mediate gonadotrophin-induced progesterone secretion, while in the testes, both Sertoli and Leydig cells express purinoceptors that mediate secretion of oestradiol and testosterone, respectively. ATP released as a cotransmitter with noradrenaline is involved in activities of the pineal gland and in the neuroendocrine control of the thymus. In the hypothalamus, ATP and adenosine stimulate or modulate the release of luteinising hormone-releasing hormone, as well as arginine-vasopressin and oxytocin. Functionally active P2X and P2Y receptors have been identified on human placental syncytiotrophoblast cells and on neuroendocrine cells in the lung, skin, prostate and intestine. Adipocytes have been recognised recently to have endocrine function involving purinoceptors.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF, UK,
| |
Collapse
|
37
|
Eisenstein A, Ravid K. G protein-coupled receptors and adipogenesis: a focus on adenosine receptors. J Cell Physiol 2014; 229:414-21. [PMID: 24114647 DOI: 10.1002/jcp.24473] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 09/12/2013] [Indexed: 12/20/2022]
Abstract
G-protein coupled receptors (GPCRs) are a large family of proteins that coordinate extracellular signals to produce physiologic outcomes. Adenosine receptors (AR) are one class of GPCRs that have been shown to regulate functions as diverse as inflammation, blood flow, and cellular differentiation. Adenosine signals through four GPCRs that either inhibit (A1AR and A3AR) or activate (A2aAR and A2bAR) adenylyl cyclase. This review will focus on the role of GPCRs, and in particular, adenosine receptors, in adipogenesis. Preadipocytes differentiate to mature adipocytes as the adipose tissue expands to compensate for the consumption of excess nutrients. These newly generated adipocytes contribute to maintaining metabolic homeostasis. Understanding the key drivers of this differentiation process can aid the development of therapeutics to combat the growing obesity epidemic and associated metabolic consequences. Although much literature has covered the transcriptional events that culminate in the formation of an adipocyte, less focus has been on receptor-mediated extracellular signals that direct this process. This review will highlight GPCRs and their downstream messengers as significant players controlling adipocyte differentiation.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | | |
Collapse
|
38
|
Yip L, Taylor C, Whiting CC, Fathman CG. Diminished adenosine A1 receptor expression in pancreatic α-cells may contribute to the pathology of type 1 diabetes. Diabetes 2013; 62:4208-19. [PMID: 24264405 PMCID: PMC3837064 DOI: 10.2337/db13-0614] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Prediabetic NOD mice exhibit hyperglucagonemia, possibly due to an intrinsic α-cell defect. Here, we show that the expression of a potential glucagon inhibitor, the adenosine A1 receptor (Adora1), is gradually diminished in α-cells of NOD mice, autoantibody-positive (AA(+)) and overtly type 1 diabetic (T1D) patients during the progression of disease. We demonstrated that islet inflammation was associated with loss of Adora1 expression through the alternative splicing of Adora1. Expression of the spliced variant (Adora1-Var) was upregulated in the pancreas of 12-week-old NOD versus age-matched NOD.B10 (non-diabetes-susceptible) control mice and was detected in the pancreas of AA(+) patients but not in control subjects or overtly diabetic patients, suggesting that inflammation drives the splicing of Adora1. We subsequently demonstrated that Adora1-Var expression was upregulated in the islets of NOD.B10 mice after exposure to inflammatory cytokines and in the pancreas of NOD.SCID mice after adoptive transfer of activated autologous splenocytes. Adora1-Var encodes a dominant-negative N-terminal truncated isoform of Adora1. The splicing of Adora1 and loss of Adora1 expression on α-cells may explain the hyperglucagonemia observed in prediabetic NOD mice and may contribute to the pathogenesis of human T1D and NOD disease.
Collapse
|
39
|
Abstract
The pancreas is an organ with a central role in nutrient breakdown, nutrient sensing and release of hormones regulating whole body nutrient homeostasis. In diabetes mellitus, the balance is broken-cells can be starving in the midst of plenty. There are indications that the incidence of diabetes type 1 and 2, and possibly pancreatogenic diabetes, is rising globally. Events leading to insulin secretion and action are complex, but there is emerging evidence that intracellular nucleotides and nucleotides are not only important as intracellular energy molecules but also as extracellular signalling molecules in purinergic signalling cascades. This signalling takes place at the level of the pancreas, where the close apposition of various cells-endocrine, exocrine, stromal and immune cells-contributes to the integrated function. Following an introduction to diabetes, the pancreas and purinergic signalling, we will focus on the role of purinergic signalling and its changes associated with diabetes in the pancreas and selected tissues/organ systems affected by hyperglycaemia and other stress molecules of diabetes. Since this is the first review of this kind, a comprehensive historical angle is taken, and common and divergent roles of receptors for nucleotides and nucleosides in different organ systems will be given. This integrated picture will aid our understanding of the challenges of the potential and currently used drugs targeted to specific organ/cells or disorders associated with diabetes.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London, NW3 2PF UK
- Department of Pharmacology, Melbourne University, Melbourne, Australia
| | - Ivana Novak
- Molecular and Integrative Physiology, Department of Biology, University of Copenhagen, August Krogh Building, Universitetsparken 13, 2100 Copenhagen Ø, Denmark
| |
Collapse
|
40
|
Amisten S, Salehi A, Rorsman P, Jones PM, Persaud SJ. An atlas and functional analysis of G-protein coupled receptors in human islets of Langerhans. Pharmacol Ther 2013; 139:359-91. [PMID: 23694765 DOI: 10.1016/j.pharmthera.2013.05.004] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/03/2013] [Indexed: 12/17/2022]
Abstract
G-protein coupled receptors (GPCRs) regulate hormone secretion from islets of Langerhans, and recently developed therapies for type-2 diabetes target islet GLP-1 receptors. However, the total number of GPCRs expressed by human islets, as well as their function and interactions with drugs, is poorly understood. In this review we have constructed an atlas of all GPCRs expressed by human islets: the 'islet GPCRome'. We have used this atlas to describe how islet GPCRs interact with their endogenous ligands, regulate islet hormone secretion, and interact with drugs known to target GPCRs, with a focus on drug/receptor interactions that may affect insulin secretion. The islet GPCRome consists of 293 GPCRs, a majority of which have unknown effects on insulin, glucagon and somatostatin secretion. The islet GPCRs are activated by 271 different endogenous ligands, at least 131 of which are present in islet cells. A large signalling redundancy was also found, with 119 ligands activating more than one islet receptor. Islet GPCRs are also the targets of a large number of clinically used drugs, and based on their coupling characteristics and effects on receptor signalling we identified 107 drugs predicted to stimulate and 184 drugs predicted to inhibit insulin secretion. The islet GPCRome highlights knowledge gaps in the current understanding of islet GPCR function, and identifies GPCR/ligand/drug interactions that might affect insulin secretion, which are important for understanding the metabolic side effects of drugs. This approach may aid in the design of new safer therapeutic agents with fewer detrimental effects on islet hormone secretion.
Collapse
Affiliation(s)
- Stefan Amisten
- Diabetes Research Group, Division of Diabetes & Nutritional Sciences, King's College London School of Medicine, London, UK.
| | | | | | | | | |
Collapse
|
41
|
Ohtani M, Oka T, Ohura K. Possible involvement of A₂A and A₃ receptors in modulation of insulin secretion and β-cell survival in mouse pancreatic islets. Gen Comp Endocrinol 2013; 187:86-94. [PMID: 23453966 DOI: 10.1016/j.ygcen.2013.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/07/2013] [Accepted: 02/12/2013] [Indexed: 10/27/2022]
Abstract
Adenosine A1, A₂A, A₂B and A₃ receptor mRNAs were found to be expressed in mouse pancreatic islets and Beta-TC6 cells but their physiological or pharmacological actions are not fully clarified. We showed that adenosine (100 μM) augmented insulin secretion by islets in the presence of either normal (5.5 mM) or a high concentration of glucose (20 mM). The augmentation of insulin secretion in the presence of high glucose was blocked by an A₂A antagonist, but not by A₂B and A₃ antagonists, while an A₁ antagonist potentiated the adenosine effect. An adenosine analogue 5'-N-ethylcarboxamidoadenosine (NECA) as well as A₁, A₂A and A₃ receptor agonists also produced stimulation. On the other hand, an A₃ agonist markedly reduced Beta-TC6 cell proliferation and the islet cell viability, while adenosine and NECA did not. The effect of A₃ agonist was partially blocked by the A₃ antagonist. In addition, treatment with the A₃ agonist produced a small but significant extent of apoptosis in Beta-TC6 cells as judged by terminal transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) assay. These results combined together suggested that like the A₁ receptor, activation of A₂A receptors by adenosine results in augmented insulin secretion, while the A₃ receptor is involved in modulation of the survival of pancreatic β-cells.
Collapse
Affiliation(s)
- M Ohtani
- Department of Pharmacology, Osaka Dental University, 8-1 Kuzuhahanazono-cho, Hirakata, Osaka 573-1121, Japan.
| | | | | |
Collapse
|
42
|
Chen JF, Eltzschig HK, Fredholm BB. Adenosine receptors as drug targets--what are the challenges? Nat Rev Drug Discov 2013; 12:265-86. [PMID: 23535933 PMCID: PMC3930074 DOI: 10.1038/nrd3955] [Citation(s) in RCA: 662] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adenosine signalling has long been a target for drug development, with adenosine itself or its derivatives being used clinically since the 1940s. In addition, methylxanthines such as caffeine have profound biological effects as antagonists at adenosine receptors. Moreover, drugs such as dipyridamole and methotrexate act by enhancing the activation of adenosine receptors. There is strong evidence that adenosine has a functional role in many diseases, and several pharmacological compounds specifically targeting individual adenosine receptors--either directly or indirectly--have now entered the clinic. However, only one adenosine receptor-specific agent--the adenosine A2A receptor agonist regadenoson (Lexiscan; Astellas Pharma)--has so far gained approval from the US Food and Drug Administration (FDA). Here, we focus on the biology of adenosine signalling to identify hurdles in the development of additional pharmacological compounds targeting adenosine receptors and discuss strategies to overcome these challenges.
Collapse
Affiliation(s)
- Jiang-Fan Chen
- Department of Neurology and Pharmacology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | |
Collapse
|
43
|
Koupenova M, Ravid K. Adenosine, adenosine receptors and their role in glucose homeostasis and lipid metabolism. J Cell Physiol 2013; 228:1703-1712. [PMID: 23460239 PMCID: PMC3849123 DOI: 10.1002/jcp.24352] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2011] [Revised: 02/09/2013] [Accepted: 02/12/2013] [Indexed: 01/12/2023]
Abstract
Adenosine is an endogenous metabolite that is released from all tissues and cells including liver, pancreas, muscle and fat, particularly under stress, intense exercise, or during cell damage. The role of adenosine in glucose homeostasis has been attributed to its ability to regulate, through its membrane receptors, processes such as insulin secretion, glucose release and clearance, glycogenolysis, and glycogenesis. Additionally, adenosine and its multiple receptors have been connected to lipid metabolism by augmenting insulin-mediated inhibition of lipolysis, and the subsequent increase in free fatty acids and glycerol levels. Furthermore, adenosine was reported to control liver cholesterol synthesis, consequently affecting plasma levels of cholesterol and triglycerides, and the amount of fat tissue. Alterations in the balance of glucose and lipid homeostasis have implications in both cardiovascular disease and diabetes. The ability of different adenosine receptors to activate and inhibit the same signaling cascades has made it challenging to study the influence of adenosine, adenosine analogs and their receptors in health and disease. This review focuses on the role and significance of different adenosine receptors in mediating the effect of adenosine on glucose and lipid homeostasis. J. Cell. Physiol. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Milka Koupenova
- Department of Medicine, Boston University School of Medicine, Boston, MA; Department of Biochemistry, Boston University School of Medicine, Boston, MA; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA
| | | |
Collapse
|
44
|
The CD39-adenosinergic axis in the pathogenesis of immune and nonimmune diabetes. J Biomed Biotechnol 2012; 2012:320495. [PMID: 23118504 PMCID: PMC3480695 DOI: 10.1155/2012/320495] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/27/2012] [Indexed: 11/24/2022] Open
Abstract
Diabetes mellitus encompasses two distinct disease processes: autoimmune Type 1 (T1D) and nonimmune Type 2 (T2D) diabetes. Despite the disparate aetiologies, the disease phenotype of hyperglycemia and the associated complications are similar. In this paper, we discuss the role of the CD39-adenosinergic axis in the pathogenesis of both T1D and T2D, with particular emphasis on the role of CD39 and CD73.
Collapse
|
45
|
Yang GK, Fredholm BB, Kieffer TJ, Kwok YN. Improved blood glucose disposal and altered insulin secretion patterns in adenosine A(1) receptor knockout mice. Am J Physiol Endocrinol Metab 2012; 303:E180-90. [PMID: 22550063 DOI: 10.1152/ajpendo.00050.2012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by the inability of the pancreatic β-cells to secrete enough insulin to meet the demands of the body. Therefore, research of potential therapeutic approaches to treat T2DM has focused on increasing insulin output from β-cells or improving systemic sensitivity to circulating insulin. In this study, we examined the role of the A(1) receptor in glucose homeostasis with the use of A(1) receptor knockout mice (A(1)R(-/-)). A(1)R(-/-) mice exhibited superior glucose tolerance compared with wild-type controls. However, glucose-stimulated insulin release, insulin sensitivity, weight gain, and food intake were comparable between the two genotypes. Following a glucose challenge, plasma glucagon levels in wild-type controls decreased, but this was not observed in A(1)R(-/-) mice. In addition, pancreas perfusion with oscillatory glucose levels of 10-min intervals produced a regular pattern of pulsatile insulin release with a 10-min cycling period in wild-type controls and 5 min in A(1)R(-/-) mice. When the mice were fed a high-fat diet (HFD), both genotypes exhibited impaired glucose tolerance and insulin resistance. Increased insulin release was observed in HFD-fed mice in both genotypes, but increased glucagon release was observed only in HFD-fed A(1)R(-/-) mice. In addition, the regular patterns of insulin release following oscillatory glucose perfusion were abolished in HFD-fed mice in both genotypes. In conclusion, A(1) receptors in the pancreas are involved in regulating the temporal patterns of insulin release, which could have implications in the development of glucose intolerance seen in T2DM.
Collapse
Affiliation(s)
- Gary K Yang
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
46
|
Koupenova M, Johnston-Cox H, Vezeridis A, Gavras H, Yang D, Zannis V, Ravid K. A2b adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation 2011; 125:354-63. [PMID: 22144568 DOI: 10.1161/circulationaha.111.057596] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The cAMP-elevating A(2b) adenosine receptor (A(2b)AR) controls inflammation via its expression in bone marrow cells. METHODS AND RESULTS Atherosclerosis induced by a high-fat diet in apolipoprotein E-deficient mice was more pronounced in the absence of the A(2b)AR. Bone marrow transplantation experiments indicated that A(2b)AR bone marrow cell signals alone were not sufficient to elicit this effect. Intriguingly, liver expression of the A(2b)AR in wild-type mice was vastly augmented by a high-fat diet, raising the possibility that this upregulation is of functional significance. A(2b)AR genetic ablation led to elevated levels of liver and plasma cholesterol and triglycerides and to fatty liver pathology typical of steatosis, assessed by enzymatic assays and analysis of liver sections. Western blotting and quantitative polymerase chain reaction revealed elevated expression of the following molecules in the liver of A(2b)AR-null mice: the transcription factor sterol regulatory element binding protein-1 (SREBP-1) and its 2 downstream targets and regulators of lipogenesis, acetyl CoA carboxylase and fatty acid synthase. Pharmacological activation or inhibition of A(2b)AR in primary hepatocytes confirmed the regulation of SREBP-1 by this receptor. A(2b)AR-mediated changes in cAMP were found to regulate levels of the transcriptionally active form of SREBP-1. Finally, adenovirally mediated restoration of the A(2b)AR in the liver of A(2b)AR-null mice reduced the lipid profile and atherosclerosis. Similarly, in vivo administration of the A(2b)AR ligand BAY 60-6853 in control mice on a high-fat diet reduced the lipid profile and atherosclerosis. CONCLUSION This study provides the first evidence that the A(2b)AR regulates liver SREBP-1, hyperlipidemia, and atherosclerosis, suggesting that this receptor may be an effective therapeutic target.
Collapse
Affiliation(s)
- Milka Koupenova
- Department of Medicine, Boston University School of Medicine, 700 Albany St, CVI, W-601, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Zywert A, Szkudelska K, Szkudelski T. Effects of adenosine A(1) receptor antagonism on insulin secretion from rat pancreatic islets. Physiol Res 2011; 60:905-11. [PMID: 21995904 DOI: 10.33549/physiolres.932165] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Adenosine is known to influence different kinds of cells, including beta-cells of the pancreas. However, the role of this nucleoside in the regulation of insulin secretion is not fully elucidated. In the present study, the effects of adenosine A(1) receptor antagonism on insulin secretion from isolated rat pancreatic islets were tested using DPCPX, a selective adenosine A(1) receptor antagonist. It was demonstrated that pancreatic islets stimulated with 6.7 and 16.7 mM glucose and exposed to DPCPX released significantly more insulin compared with islets incubated with glucose alone. The insulin-secretory response to glucose and low forskolin appeared to be substantially potentiated by DPCPX, but DPCPX was ineffective in the presence of glucose and high forskolin. Moreover, DPCPX failed to change insulin secretion stimulated by the combination of glucose and dibutyryl-cAMP, a non-hydrolysable cAMP analogue. Studies on pancreatic islets also revealed that the potentiating effect of DPCPX on glucose-induced insulin secretion was attenuated by H-89, a selective inhibitor of protein kinase A. It was also demonstrated that formazan formation, reflecting metabolic activity of cells, was enhanced in islets exposed to DPCPX. Moreover, DPCPX was found to increase islet cAMP content, whereas ATP was not significantly changed. These results indicate that adenosine A(1) receptor blockade in rat pancreatic islets potentiates insulin secretion induced by both physiological and supraphysiological glucose concentrations. This effect is proposed to be due to increased metabolic activity of cells and increased cAMP content.
Collapse
Affiliation(s)
- A Zywert
- Department of Animal Physiology and Biochemistry, Poznan University of Life Sciences, Poznan, Poland
| | | | | |
Collapse
|
48
|
Faulhaber-Walter R, Jou W, Mizel D, Li L, Zhang J, Kim SM, Huang Y, Chen M, Briggs JP, Gavrilova O, Schnermann JB. Impaired glucose tolerance in the absence of adenosine A1 receptor signaling. Diabetes 2011; 60:2578-87. [PMID: 21831968 PMCID: PMC3178298 DOI: 10.2337/db11-0058] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The role of adenosine (ADO) in the regulation of glucose homeostasis is not clear. In the current study, we used A1-ADO receptor (A1AR)-deficient mice to investigate the role of ADO/A1AR signaling for glucose homeostasis. RESEARCH DESIGN AND METHODS After weaning, A1AR(-/-) and wild-type mice received either a standard diet (12 kcal% fat) or high-fat diet (HFD; 45 kcal% fat). Body weight, fasting plasma glucose, plasma insulin, and intraperitoneal glucose tolerance tests were performed in 8-week-old mice and again after 12-20 weeks of subsequent observation. Body composition was quantified by magnetic resonance imaging and epididymal fat-pad weights. Glucose metabolism was investigated by hyperinsulinemic-euglycemic clamp studies. To describe pathophysiological mechanisms, adipokines and Akt phosphorylation were measured. RESULTS A1AR(-/-) mice were significantly heavier than wild-type mice because of an increased fat mass. Fasting plasma glucose and insulin were significantly higher in A1AR(-/-) mice after weaning and remained higher in adulthood. An intraperitoneal glucose challenge disclosed a significantly slower glucose clearance in A1AR(-/-) mice. An HFD enhanced this phenotype in A1AR(-/-) mice and unmasked a dysfunctional insulin secretory mechanism. Insulin sensitivity was significantly impaired in A1AR(-/-) mice on the standard diet shortly after weaning. Clamp studies detected a significant decrease of net glucose uptake in A1AR(-/-) mice and a reduced glucose uptake in muscle and white adipose tissue. Effects were not triggered by leptin deficiency but involved a decreased Akt phosphorylation. CONCLUSIONS ADO/A1AR signaling contributes importantly to insulin-controlled glucose homeostasis and insulin sensitivity in C57BL/6 mice and is involved in the metabolic regulation of adipose tissue.
Collapse
Affiliation(s)
- Robert Faulhaber-Walter
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Gharibi B, Abraham AA, Ham J, Evans BAJ. Contrasting effects of A1 and A2b adenosine receptors on adipogenesis. Int J Obes (Lond) 2011; 36:397-406. [PMID: 21730968 DOI: 10.1038/ijo.2011.129] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Adenosine mediates its actions through four G protein-coupled receptors, A1, A2a, A2b and A3. The A1 receptor (A1R) is dominant in adipocytes where it mediates many actions that include inhibition of lipolysis, stimulation of leptin secretion and protection against obesity-related insulin resistance. OBJECTIVE The objective of this study is to investigate whether induced expression of A1Rs stimulates adipogenesis, or whether A1R expression is a consequence of cells having an adipocyte phenotype. METHODOLOGY Human A1R and A2b receptors (A2bRs) were stably transfected into a murine osteoblast precursor cell line, 7F2. Adipogenesis was determined by lipid accumulation and expression of adipocyte and osteoblast marker molecules. Adenosine receptor expression and activation of associated signal molecules were also evaluated as 7F2 cells were induced to differentiate to adipocytes. RESULTS 7F2 cells transfected with the A1R showed increased adipocyte marker mRNA expression; lipoprotein lipase and glycerol-3-phosphate dehydrogenase were both upregulated, whereas the osteoblast marker alkaline phosphatase (ALP) was downregulated. When cultured in adipocyte differentiating media, such cells also showed increased adipogenesis as judged by lipid accumulation. Conversely, A2bR transfection stimulated osteocalcin and ALP expression, and in addition, adipogenesis was inhibited in the presence of adipocyte differentiation media. Adipogenic differentiation of naive 7F2 cells also resulted in increased expression of the A1R and reduced or modified expression of the A2a and A2bR. The loss of A2 receptors after adipogenic differentiation was accompanied by a loss of cyclic adenosine monophosphate and ERK1/2 signalling. CONCLUSION These data show that expression of A1Rs induced adipocyte differentiation, whereas A2bR expression inhibited adipogenesis and stimulated an osteoblastic phenotype. These data suggest that targeting A1 and A2bR could be considered in the management of obesity and diabetes. Targeting adenosine signal pathways may be useful in treatment strategies for diseases in which there is an imbalance between osteoblasts and adipocytes.
Collapse
Affiliation(s)
- B Gharibi
- Centre for Endocrine and Diabetes Sciences, School of Medicine, Cardiff University, Heath Park, Cardiff, UK
| | | | | | | |
Collapse
|
50
|
Fredholm BB, IJzerman AP, Jacobson KA, Linden J, Müller CE. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors--an update. Pharmacol Rev 2011; 63:1-34. [PMID: 21303899 PMCID: PMC3061413 DOI: 10.1124/pr.110.003285] [Citation(s) in RCA: 1032] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the 10 years since our previous International Union of Basic and Clinical Pharmacology report on the nomenclature and classification of adenosine receptors, no developments have led to major changes in the recommendations. However, there have been so many other developments that an update is needed. The fact that the structure of one of the adenosine receptors has recently been solved has already led to new ways of in silico screening of ligands. The evidence that adenosine receptors can form homo- and heteromultimers has accumulated, but the functional significance of such complexes remains unclear. The availability of mice with genetic modification of all the adenosine receptors has led to a clarification of the functional roles of adenosine, and to excellent means to study the specificity of drugs. There are also interesting associations between disease and structural variants in one or more of the adenosine receptors. Several new selective agonists and antagonists have become available. They provide improved possibilities for receptor classification. There are also developments hinting at the usefulness of allosteric modulators. Many drugs targeting adenosine receptors are in clinical trials, but the established therapeutic use is still very limited.
Collapse
Affiliation(s)
- Bertil B Fredholm
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|