1
|
Luca BGD, Almeida PP, Junior RR, Soares DJS, Frantz EDC, Miranda-Alves L, Stockler-Pinto MB, Machado Dos Santos C, Magliano DC. Environmental contamination by bisphenols: From plastic production to modulation of the intestinal morphophysiology in experimental models. Food Chem Toxicol 2025; 197:115280. [PMID: 39923829 DOI: 10.1016/j.fct.2025.115280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 02/11/2025]
Abstract
Bisphenols are frequently found in a range of plastic products and have been associated with the development of diseases such as diabetes mellitus type 2 and obesity. These compounds are known as endocrine disruptors and have led to restrictions on their use due to their presence in the environment and their association with non-communicable chronic diseases. The gastrointestinal tract, being the primary site of food and water absorption, is particularly vulnerable to the effects of bisphenols. For this reason, a review of studies showing associations between bisphenols exposure and adverse effects in the gut microbiota, morphology tissue, gut permeability, and on the enteric nervous system was carried out. We have included perinatal studies and in different adult experimental models. The effects of bisphenol exposure on the gut microbiota are complex and varied. Bisphenol exposure generally leads to a decrease in microbial diversity and may impact the integrity of the intestinal barrier, resulting in elevated levels of inflammation, changes in morphological and metabolic characteristics of the gut, modifications in tight junction expression, and changes in goblet cell expression. In addition, bisphenol exposure in the perinatal phase can lead to important intestinal changes, including increased colonic inflammation and decreased colonic paracellular permeability.
Collapse
Affiliation(s)
- Beatriz Gouvêa de Luca
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Laboratory of Teaching and Research in Histology and Comparative Embryology (LEPHEC), Federal Fluminense University, Niterói, RJ, Brazil; Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Patricia Pereira Almeida
- Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Nutrition Sciences Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Reinaldo Röpke Junior
- Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil
| | - Débora Júlia Silva Soares
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil
| | - Eliete Dalla Corte Frantz
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Cardiovascular Sciences Graduate Program, Fluminense Federal University (UFF), Niteroi, RJ, Brazil
| | - Leandro Miranda-Alves
- Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil; Pharmacology and Medicinal Chemistry Graduate Program, Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Brazil; Morphological Sciences Graduate Program, Institute of Biomedical Sciences, Federal University of Rio de Janeiro (UFRJ), Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Nutrition Sciences Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil
| | - Clarice Machado Dos Santos
- Laboratory of Teaching and Research in Histology and Comparative Embryology (LEPHEC), Federal Fluminense University, Niterói, RJ, Brazil
| | - D'Angelo Carlo Magliano
- Research Center on Morphology and Metabolism, Biomedical Institute, Federal Fluminense University, Niteroi, RJ, Brazil; Pathology Graduate Program, Federal Fluminense University (UFF), Niteroi, RJ, Brazil; Laboratory of Experimental Endocrinology (LEEx), Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Endocrinology Graduate Program, Faculty of Medicine, Federal University of Rio de Janeiro (UFRJ), Brazil.
| |
Collapse
|
2
|
Zhou Y, Kang L, Yin G, Yang L, Chen B, Liu B, Zhu X, Xie Q. Adenosine A2B receptor activation regulates the balance between T helper 17 cells and regulatory T cells, and inhibits regulatory T cells exhaustion in experimental autoimmune myositis. J Cachexia Sarcopenia Muscle 2024; 15:2460-2475. [PMID: 39284778 PMCID: PMC11634480 DOI: 10.1002/jcsm.13581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 07/11/2024] [Accepted: 07/23/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Idiopathic inflammatory myopathy (IIM) is a systemic autoimmune disease characterized by skeletal muscle involvement. This study aimed to investigate the role of adenosine receptor signalling pathways in the development of experimental autoimmune myositis (EAM). METHODS An ecto-5'-nucleotidase (CD73) inhibitor, adenosine receptor agonists, a hypoxia-inducible factor-1α (HIF-1α) inhibitor or a vehicle were administered to control and EAM mice. Murine splenic CD4+ or regulatory T cells (Tregs) were isolated using magnetic beads and subsequently stimulated with an adenosine A2B receptor agonist, a HIF-1α inhibitor, or vehicle in vitro. In cross-sectional studies, we collected 64 serum samples (69% female, 49 ± 9 years), 63 peripheral blood samples (70% female, 50 ± 11 years), and 34 skeletal muscle samples (71% female, 63 ± 6 years) from patients with IIM. Additionally, 35 serum samples and 30 peripheral blood samples were obtained from age- and sex-matched healthy controls, and six quadriceps muscle samples were collected from patients with osteoarthritis to serve as the normal group. RESULTS Patients with IIM exhibited increased CD73 [dermatomyositis (DM), polymyositis (PM): P < 0.01; immune-mediated necrotizing myopathy (IMNM): P < 0.0001] and adenosine deaminase (ADA) expression (DM: P < 0.001; PM, IMNM: P < 0.0001) in the skeletal muscles, and serum ADA levels [56.7 (95% CI: 53.7, 58.7) vs. 198.8 (95% CI: 186.2, 237.3) ng/μL, P < 0.0001]. Intervention with a CD73 inhibitor exacerbated (P = 0.0461), whereas adenosine receptor agonists (A1: P = 0.0009; A2B: P < 0.0001; A3: P = 0.0001) and the HIF-1α inhibitor (P = 0.0044) alleviated skeletal muscle injury in EAM mice. Elevated expression of programmed cell death protein-1 (PD1: P = 0.0023) and T-cell immunoglobulin and mucin-domain containing-3 (TIM3: P < 0.0001) in skeletal muscles of patients with IIM were correlated with creatine kinase levels (PD1, r = 0.7072, P < 0.0001; TIM3, r = 0.4808, P = 0.0046). PD1+CD4+ (r = 0.3243, P = 0.0115) and PD1+CD8+ (r = 0.3959, P = 0.0017) T cells were correlated with Myositis Disease Activity Assessment Visual Analogue Scale scores (muscle) in IIM. The exhausted Tregs were identified in the skeletal muscles of patients with IIM. Activation of the A2B adenosine receptor downregulated HIF-1α (protein or mRNA level, P < 0.01), resulting in decreased T helper cell 17 (Th17) (13.58% vs. 5.43%, P = 0.0201) and phosphorylated-signal transducer and activator of transcription 3 (p-STAT3)+ Th17 (16.32% vs. 6.73%, P = 0.0029), decreased exhausted Tregs (PD1+ Tregs: 53.55% vs. 40.28%, P = 0.0005; TIM3+ Tregs: 3.93% vs. 3.11%, P = 0.0029), and increased Tregs (0.45% vs. 2.89%, P = 0.0006) in EAM mice. CONCLUSIONS The exhausted T cells may be pathogenic in IIM, and the activation of adenosine A2B receptor signalling pathway can regulate Th17/Treg balance and inhibit Tregs exhaustion, thereby slowing EAM disease progression.
Collapse
Affiliation(s)
- Yueyuan Zhou
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Limei Kang
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Geng Yin
- Department of General Practice, General Practice Medical CenterWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Leiyi Yang
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Bo Chen
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Binhan Liu
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| | - Xiaoyan Zhu
- Department of PhysiologyNaval Medical UniversityShanghaiYangpu DistrictChina
| | - Qibing Xie
- Department of Rheumatology and ImmunologyWest China Hospital, Sichuan UniversityChengduWuhou DistrictChina
| |
Collapse
|
3
|
Almeida PP, Brito ML, Thomasi B, Mafra D, Fouque D, Knauf C, Tavares-Gomes AL, Stockler-Pinto MB. Is the enteric nervous system a lost piece of the gut-kidney axis puzzle linked to chronic kidney disease? Life Sci 2024; 351:122793. [PMID: 38848938 DOI: 10.1016/j.lfs.2024.122793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
The enteric nervous system (ENS) regulates numerous functional and immunological attributes of the gastrointestinal tract. Alterations in ENS cell function have been linked to intestinal outcomes in various metabolic, intestinal, and neurological disorders. Chronic kidney disease (CKD) is associated with a challenging intestinal environment due to gut dysbiosis, which further affects patient quality of life. Although the gut-related repercussions of CKD have been thoroughly investigated, the involvement of the ENS in this puzzle remains unclear. ENS cell dysfunction, such as glial reactivity and alterations in cholinergic signaling in the small intestine and colon, in CKD are associated with a wide range of intestinal pathways and responses in affected patients. This review discusses how the ENS is affected in CKD and how it is involved in gut-related outcomes, including intestinal permeability, inflammation, oxidative stress, and dysmotility.
Collapse
Affiliation(s)
| | - Michele Lima Brito
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Beatriz Thomasi
- Department of Physiology, Neuroscience Program, Michigan State University (MSU), East Lansing, MI, USA
| | - Denise Mafra
- Graduate Program in Biological Sciences - Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Denis Fouque
- Department of Nephrology, Centre Hopitalier Lyon Sud, INSERM 1060, CENS, Université de Lyon, France
| | - Claude Knauf
- INSERM U1220 Institut de Recherche en Santé Digestive, CHU Purpan, Université Toulouse III Paul Sabatier Toulouse, Toulouse, France
| | - Ana Lúcia Tavares-Gomes
- Neurosciences Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Milena Barcza Stockler-Pinto
- Pathology Post Graduate Program, Fluminense Federal University (UFF), Niterói, RJ, Brazil; INSERM U1220 Institut de Recherche en Santé Digestive, CHU Purpan, Université Toulouse III Paul Sabatier Toulouse, Toulouse, France
| |
Collapse
|
4
|
6-OHDA-Induced Changes in Colonic Segment Contractility in the Rat Model of Parkinson's Disease. Gastroenterol Res Pract 2023; 2023:9090524. [PMID: 36743531 PMCID: PMC9897937 DOI: 10.1155/2023/9090524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 12/20/2022] [Accepted: 01/04/2023] [Indexed: 01/29/2023] Open
Abstract
Background Gastrointestinal dysfunction is one of the most common non-motor symptoms in Parkinson's disease (PD). The exact mechanisms behind these symptoms are not clearly understood. Studies in the well-established 6-hydroxydopamine (6-OHDA) lesioned rats of PD have shown altered contractility in isolated circular and longitudinal smooth muscle strips of distal colon. Contractile changes in proximal colon and distal ileum are nevertheless poorly studied. Moreover, segments may serve as better tissue preparations to understand the interplay between circular and longitudinal smooth muscle. This study aimed to compare changes in contractility between isolated full-thickness distal colon muscle strips and segments, and extend the investigation to proximal colon and distal ileum in the 6-OHDA rat model. Methods Spontaneous contractions and contractions induced by electrical field stimulation (EFS) and by the non-selective muscarinic agonist methacholine were investigated in strip and/or segment preparations of smooth muscle tissue from distal and proximal colon and distal ileum in an in vitro organ bath comparing 6-OHDA-lesioned rats with Sham-operated animals. Key Results. Our data showed increased contractility evoked by EFS and methacholine in segments, but not in circular and longitudinal tissue strips of distal colon after central 6-OHDA-induced dopamine denervation. Changes in proximal colon segments were also displayed in high K+ Krebs-induced contractility and spontaneous contractions. Conclusions This study further confirms changes in smooth muscle contractility in distal colon and to some extent in proximal colon, but not in distal ileum in the 6-OHDA rat model of PD. However, the changes depended on tissue preparation.
Collapse
|
5
|
D'Antongiovanni V, Pellegrini C, Benvenuti L, Fornai M, Di Salvo C, Natale G, Ryskalin L, Bertani L, Lucarini E, Di Cesare Mannelli L, Ghelardini C, Nemeth ZH, Haskó G, Antonioli L. Anti-inflammatory Effects of Novel P2X4 Receptor Antagonists, NC-2600 and NP-1815-PX, in a Murine Model of Colitis. Inflammation 2022; 45:1829-1847. [PMID: 35338432 PMCID: PMC9197920 DOI: 10.1007/s10753-022-01663-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022]
Abstract
The pharmacological blockade of P2X4 receptors has shown potential benefits in the management of several immune/inflammatory diseases. However, data regarding the involvement of P2X4 receptors in the pathophysiological mechanisms of action in intestinal inflammation are not well defined. We aimed to evaluate the anti-inflammatory effects of two novel and selective P2X4 receptor antagonists, NC-2600 and NP-1815-PX, and characterize the molecular mechanisms of their action in a murine model of 2,4-dinitrobenzene sulfonic acid (DNBS)-induced colitis. These two drugs and dexamethasone (DEX) were administered orally for 6 days, immediately after the manifestation of DNBS. The body weight decrease, resulting from colitis, was attenuated by NC-2600 and NP-1815-PX, but not DEX. However, all three drugs attenuated the increase in spleen weight and ameliorated macroscopic and microscopic colonic tissue damage. Furthermore, all three compounds decreased tissue IL-1β levels and caspase-1 expression and activity. Colonic tissue increase of tumor necrosis factor was downregulated by DEX, while both NC-2600 and NP-1815-PX were ineffective. The reduction of occludin associated with colitis was ameliorated by NC-2600 and NP-1815-PX, but not DEX. In THP-1 cells, lipopolysaccharide and ATP upregulated IL-1β release and NLRP3, caspase-1, caspase-5, and caspase-8 activity, but not of caspase-4. These changes were prevented by NC-2600 and NP-1815-PX treatment. For the first time, the above findings show that the selective inhibition of P2X4 receptors represents a viable approach to manage bowel inflammation via the inhibition of NLRP3 inflammasome signaling pathways.
Collapse
Affiliation(s)
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa Via Roma 55, 56126 Pisa, Italy.
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Zoltan H Nemeth
- Department of Surgery, Morristown Medical Center, Morristown, NJ, 07960, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
6
|
Antonioli L, Pellegrini C, Fornai M, Benvenuti L, D’Antongiovanni V, Colucci R, Bertani L, Di Salvo C, Semeghini G, La Motta C, Giusti L, Zallocco L, Ronci M, Quattrini L, Angelucci F, Coviello V, Oh WK, Ha QTK, Németh ZH, Haskó G, Blandizzi C. Preclinical Development of FA5, a Novel AMP-Activated Protein Kinase (AMPK) Activator as an Innovative Drug for the Management of Bowel Inflammation. Int J Mol Sci 2021; 22:6325. [PMID: 34199160 PMCID: PMC8231528 DOI: 10.3390/ijms22126325] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
Acadesine (ACA), a pharmacological activator of AMP-activated protein kinase (AMPK), showed a promising beneficial effect in a mouse model of colitis, indicating this drug as an alternative tool to manage IBDs. However, ACA displays some pharmacodynamic limitations precluding its therapeutical applications. Our study was aimed at evaluating the in vitro and in vivo effects of FA-5 (a novel direct AMPK activator synthesized in our laboratories) in an experimental model of colitis in rats. A set of experiments evaluated the ability of FA5 to activate AMPK and to compare the efficacy of FA5 with ACA in an experimental model of colitis. The effects of FA-5, ACA, or dexamethasone were tested in rats with 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis to assess systemic and tissue inflammatory parameters. In in vitro experiments, FA5 induced phosphorylation, and thus the activation, of AMPK, contextually to the activation of SIRT-1. In vivo, FA5 counteracted the increase in spleen weight, improved the colon length, ameliorated macroscopic damage score, and reduced TNF and MDA tissue levels in DNBS-treated rats. Of note, FA-5 displayed an increased anti-inflammatory efficacy as compared with ACA. The novel AMPK activator FA-5 displays an improved anti-inflammatory efficacy representing a promising pharmacological tool against bowel inflammation.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Carolina Pellegrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Vanessa D’Antongiovanni
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy;
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy;
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Giorgia Semeghini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| | - Concettina La Motta
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Laura Giusti
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | - Lorenzo Zallocco
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Maurizio Ronci
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Luca Quattrini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Francesco Angelucci
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Vito Coviello
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (C.P.); (C.L.M.); (L.Z.); (L.Q.); (F.A.); (V.C.)
| | - Won-Keun Oh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Quy Thi Kim Ha
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Korea; (W.-K.O.); (Q.T.K.H.)
| | - Zoltan H. Németh
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
- Department of Surgery, Morristown Medical Center, Morristown, NJ 07960, USA
| | - Gyorgy Haskó
- Department of Anesthesiology, Columbia University, New York City, NY 10027, USA; (Z.H.N.); (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.F.); (L.B.); (V.D.); (C.D.S.); (G.S.); (C.B.)
| |
Collapse
|
7
|
Magni G, Ceruti S. Adenosine Signaling in Autoimmune Disorders. Pharmaceuticals (Basel) 2020; 13:ph13090260. [PMID: 32971792 PMCID: PMC7558305 DOI: 10.3390/ph13090260] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
The molecular components of the purinergic system (i.e., receptors, metabolizing enzymes and membrane transporters) are widely expressed in the cells of the immune system. Additionally, high concentrations of adenosine are generated from the hydrolysis of ATP in any "danger" condition, when oxygen and energy availability dramatically drops. Therefore, adenosine acts as a retaliatory metabolite to counteract the nucleotide-mediated boost of the immune reaction. Based on this observation, it can be foreseen that the recruitment with selective agonists of the receptors involved in the immunomodulatory effect of adenosine might represent an innovative anti-inflammatory approach with potential exploitation in autoimmune disorders. Quite surprisingly, pro-inflammatory activity exerted by some adenosine receptors has been also identified, thus paving the way for the hypothesis that at least some autoimmune disorders may be caused by a derailment of adenosine signaling. In this review article, we provide a general overview of the roles played by adenosine on immune cells with a specific focus on the development of adenosine-based therapies for autoimmune disorders, as demonstrated by the exciting data from concluded and ongoing clinical trials.
Collapse
|
8
|
Antonioli L, Fornai M, Pellegrini C, Bertani L, Nemeth ZH, Blandizzi C. Inflammatory Bowel Diseases: It's Time for the Adenosine System. Front Immunol 2020; 11:1310. [PMID: 32849492 PMCID: PMC7403190 DOI: 10.3389/fimmu.2020.01310] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 05/22/2020] [Indexed: 12/16/2022] Open
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Zoltan H Nemeth
- Department of Surgery, Morristown Medical Center, Morristown, NJ, United States.,Department of Anesthesiology, Columbia University Medical Center, New York, NY, United States
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
9
|
Shakya AK, Naik RR, Almasri IM, Kaur A. Role and Function of Adenosine and its Receptors in Inflammation, Neuroinflammation, IBS, Autoimmune Inflammatory Disorders, Rheumatoid Arthritis and Psoriasis. Curr Pharm Des 2020; 25:2875-2891. [PMID: 31333103 DOI: 10.2174/1381612825666190716145206] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
The physiological effects of endogenous adenosine on various organ systems are very complex and numerous which are elicited upon activation of any of the four G-protein-coupled receptors (GPCRs) denoted as A1, A2A, A2B and A3 adenosine receptors (ARs). Several fused heterocyclic and non-xanthine derivatives are reported as a possible target for these receptors due to physiological problems and lack of selectivity of xanthine derivatives. In the present review, we have discussed the development of various new chemical entities as a target for these receptors. In addition, compounds acting on adenosine receptors can be utilized in treating diseases like inflammation, neuroinflammation, autoimmune and related diseases.
Collapse
Affiliation(s)
- Ashok K Shakya
- Medicinal Chemistry, Drug Design and Drug Metabolism, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al- Ahliyya Amman University, PO Box 263, Amman 19328, Jordan
| | - Rajashri R Naik
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Ihab M Almasri
- Medicinal Chemistry and Drug Design, Department of Pharmaceutical Chemistry and Pharmacognosy, Faculty of Pharmacy, Al Azhar University Gaza, Gaza Strip, Palestinian Territory, Occupied
| | - Avneet Kaur
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), Mehrauli-Badarpur Road, Pushp Vihar, Sector-3, New Delhi-110017, India
| |
Collapse
|
10
|
Asano T, Noda Y, Tanaka KI, Yamakawa N, Wada M, Mashimo T, Fukunishi Y, Mizushima T, Takenaga M. A 2B adenosine receptor inhibition by the dihydropyridine calcium channel blocker nifedipine involves colonic fluid secretion. Sci Rep 2020; 10:3555. [PMID: 32103051 PMCID: PMC7044278 DOI: 10.1038/s41598-020-60147-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 01/28/2020] [Indexed: 11/26/2022] Open
Abstract
The adenosine A2B receptor is a critical protein in intestinal water secretion. In the present study, we screened compound libraries to identify inhibitors of the A2B receptor and evaluated their effect on adenosine-induced intestinal fluid secretion. The screening identified the dihydropyridine calcium antagonists nifedipine and nisoldipine. Their respective affinities for the A2B receptor (Ki value) were 886 and 1,399 nM. Nifedipine and nisoldipine, but not amlodipine or nitrendipine, inhibited both calcium mobilization and adenosine-induced cAMP accumulation in cell lines. Moreover, adenosine injection into the lumen significantly increased fluid volume in the colonic loop of wild-type mice but not A2B receptor-deficient mice. PSB-1115, a selective A2B receptor antagonist, and nifedipine prevented elevated adenosine-stimulated fluid secretion in mice. Our results may provide useful insights into the structure–activity relationship of dihydropyridines for A2B receptor. As colonic fluid secretion by adenosine seems to rely predominantly on the A2B receptor, nifedipine could be a therapeutic candidate for diarrhoea-related diseases.
Collapse
Affiliation(s)
- Teita Asano
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, 216-8512, Japan.
| | - Yuto Noda
- LTT Bio-Pharma Co., Ltd, Shiodome Building 3F, 1-2-20 Kaigan, Minato-ku, Tokyo, 105-0022, Japan
| | - Ken-Ichiro Tanaka
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20, Shin-machi, Nishi-Tokyo, 202-8585, Japan
| | - Naoki Yamakawa
- School of Pharmacy, Shujitsu University, 1-6-1, Nishi-kawahara, Naka-ku, Okayama, 703-8516, Japan
| | - Mitsuhito Wada
- Technology Research Association for Next Generation Natural Products Chemistry, 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Tadaaki Mashimo
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan.,IMSBIO Co., Ltd., Owl Tower, 4-21-1, Higashi-Ikebukuro, Toshima-ku, Tokyo, 170-0013, Japan
| | - Yoshifumi Fukunishi
- Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-3-26, Aomi, Koto-ku, Tokyo, 135-0064, Japan
| | - Tohru Mizushima
- LTT Bio-Pharma Co., Ltd, Shiodome Building 3F, 1-2-20 Kaigan, Minato-ku, Tokyo, 105-0022, Japan.
| | - Mitsuko Takenaga
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki, 216-8512, Japan
| |
Collapse
|
11
|
Bagheri S, Saboury AA, Haertlé T. Adenosine deaminase inhibition. Int J Biol Macromol 2019; 141:1246-1257. [PMID: 31520704 DOI: 10.1016/j.ijbiomac.2019.09.078] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 12/18/2022]
Abstract
Adenosine deaminase is a critical enzyme in purine metabolism that regulates intra and extracellular adenosine concentrations by converting it to inosine. Adenosine is an important purine that regulates numerous physiological functions by interacting with its receptors. Adenosine and consequently adenosine deaminase can have pro or anti-inflammatory effects on tissues depending on how much time has passed from the start of the injury. In addition, an increase in adenosine deaminase activity has been reported for various diseases and the significant effect of deaminase inhibition on the clinical course of different diseases has been reported. However, the use of inhibitors is limited to only a few medical indications. Data on the increase of adenosine deaminase activity in different diseases and the impact of its inhibition in various cases have been collected and are discussed in this review. Overall, the evidence shows that many studies have been done to introduce inhibitors, however, in vivo studies have been much less than in vitro, and often have not been expanded for clinical use.
Collapse
Affiliation(s)
- S Bagheri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - A A Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | - T Haertlé
- Institut National de la Recherche Agronomique, Nantes, France
| |
Collapse
|
12
|
Antonioli L, Blandizzi C, Pacher P, Haskó G. The Purinergic System as a Pharmacological Target for the Treatment of Immune-Mediated Inflammatory Diseases. Pharmacol Rev 2019; 71:345-382. [PMID: 31235653 PMCID: PMC6592405 DOI: 10.1124/pr.117.014878] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) encompass a wide range of seemingly unrelated conditions, such as multiple sclerosis, rheumatoid arthritis, psoriasis, inflammatory bowel diseases, asthma, chronic obstructive pulmonary disease, and systemic lupus erythematosus. Despite differing etiologies, these diseases share common inflammatory pathways, which lead to damage in primary target organs and frequently to a plethora of systemic effects as well. The purinergic signaling complex comprising extracellular nucleotides and nucleosides and their receptors, the P2 and P1 purinergic receptors, respectively, as well as catabolic enzymes and nucleoside transporters is a major regulatory system in the body. The purinergic signaling complex can regulate the development and course of IMIDs. Here we provide a comprehensive review on the role of purinergic signaling in controlling immunity, inflammation, and organ function in IMIDs. In addition, we discuss the possible therapeutic applications of drugs acting on purinergic pathways, which have been entering clinical development, to manage patients suffering from IMIDs.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - Pál Pacher
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| | - György Haskó
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (L.A., C.B.); Laboratory of Cardiovascular Physiology and Tissue Injury, National Institutes of Health, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland (P.P.); and Department of Anesthesiology, Columbia University, New York, New York (G.H.)
| |
Collapse
|
13
|
Emerging Role of Purine Metabolizing Enzymes in Brain Function and Tumors. Int J Mol Sci 2018; 19:ijms19113598. [PMID: 30441833 PMCID: PMC6274932 DOI: 10.3390/ijms19113598] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022] Open
Abstract
The growing evidence of the involvement of purine compounds in signaling, of nucleotide imbalance in tumorigenesis, the discovery of purinosome and its regulation, cast new light on purine metabolism, indicating that well known biochemical pathways may still surprise. Adenosine deaminase is important not only to preserve functionality of immune system but also to ensure a correct development and function of central nervous system, probably because its activity regulates the extracellular concentration of adenosine and therefore its function in brain. A lot of work has been done on extracellular 5′-nucleotidase and its involvement in the purinergic signaling, but also intracellular nucleotidases, which regulate the purine nucleotide homeostasis, play unexpected roles, not only in tumorigenesis but also in brain function. Hypoxanthine guanine phosphoribosyl transferase (HPRT) appears to have a role in the purinosome formation and, therefore, in the regulation of purine synthesis rate during cell cycle with implications in brain development and tumors. The final product of purine catabolism, uric acid, also plays a recently highlighted novel role. In this review, we discuss the molecular mechanisms underlying the pathological manifestations of purine dysmetabolisms, focusing on the newly described/hypothesized roles of cytosolic 5′-nucleotidase II, adenosine kinase, adenosine deaminase, HPRT, and xanthine oxidase.
Collapse
|
14
|
The ecto-enzymes CD73 and adenosine deaminase modulate 5'-AMP-derived adenosine in myofibroblasts of the rat small intestine. Purinergic Signal 2018; 14:409-421. [PMID: 30269308 DOI: 10.1007/s11302-018-9623-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
Adenosine is a versatile signaling molecule recognized to physiologically influence gut motor functions. Both the duration and magnitude of adenosine signaling in enteric neuromuscular function depend on its availability, which is regulated by the ecto-enzymes ecto-5'-nucleotidase (CD73), alkaline phosphatase (AP), and ecto-adenosine deaminase (ADA) and by dipyridamole-sensitive equilibrative transporters (ENTs). Our purpose was to assess the involvement of CD73, APs, ecto-ADA in the formation of AMP-derived adenosine in primary cultures of ileal myofibroblasts (IMFs). IMFs were isolated from rat ileum longitudinal muscle segments by means of primary explant technique and identified by immunofluorescence staining for vimentin and α-smooth muscle actin. IMFs confluent monolayers were exposed to exogenous 5'-AMP in the presence or absence of CD73, APs, ecto-ADA, or ENTs inhibitors. The formation of adenosine and its metabolites in the IMFs medium was monitored by high-performance liquid chromatography. The distribution of CD73 and ADA in IMFs was detected by confocal immunocytochemistry and qRT-PCR. Exogenous 5'-AMP was rapidly cleared being almost undetectable after 60-min incubation, while adenosine levels significantly increased. Treatment of IMFs with CD73 inhibitors markedly reduced 5'-AMP clearance whereas ADA blockade or inhibition of both ADA and ENTs prevented adenosine catabolism. By contrast, inhibition of APs did not affect 5'-AMP metabolism. Immunofluorescence staining and qRT-PCR analysis confirmed the expression of CD73 and ADA in IMFs. Overall, our data show that in IMFs an extracellular AMP-adenosine pathway is functionally active and among the different enzymatic pathways regulating extracellular adenosine levels, CD73 and ecto-ADA represent the critical catabolic pathway.
Collapse
|
15
|
Moreno E, Canet J, Gracia E, Lluís C, Mallol J, Canela EI, Cortés A, Casadó V. Molecular Evidence of Adenosine Deaminase Linking Adenosine A 2A Receptor and CD26 Proteins. Front Pharmacol 2018; 9:106. [PMID: 29497379 PMCID: PMC5818423 DOI: 10.3389/fphar.2018.00106] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 01/30/2018] [Indexed: 01/05/2023] Open
Abstract
Adenosine is an endogenous purine nucleoside that acts in all living systems as a homeostatic network regulator through many pathways, which are adenosine receptor (AR)-dependent and -independent. From a metabolic point of view, adenosine deaminase (ADA) is an essential protein in the regulation of the total intracellular and extracellular adenosine in a tissue. In addition to its cytosolic localization, ADA is also expressed as an ecto-enzyme on the surface of different cells. Dipeptidyl peptidase IV (CD26) and some ARs act as binding proteins for extracellular ADA in humans. Since CD26 and ARs interact with ADA at opposite sites, we have investigated if ADA can function as a cell-to-cell communication molecule by bridging the anchoring molecules CD26 and A2AR present on the surfaces of the interacting cells. By combining site-directed mutagenesis of ADA amino acids involved in binding to A2AR and a modification of the bioluminescence resonance energy transfer (BRET) technique that allows detection of interactions between two proteins expressed in different cell populations with low steric hindrance (NanoBRET), we show direct evidence of the specific formation of trimeric complexes CD26-ADA-A2AR involving two cells. By dynamic mass redistribution assays and ligand binding experiments, we also demonstrate that A2AR-NanoLuc fusion proteins are functional. The existence of this ternary complex is in good agreement with the hypothesis that ADA could bridge T-cells (expressing CD26) and dendritic cells (expressing A2AR). This is a new metabolic function for ecto-ADA that, being a single chain protein, it has been considered as an example of moonlighting protein, because it performs more than one functional role (as a catalyst, a costimulator, an allosteric modulator and a cell-to-cell connector) without partitioning these functions in different subunits.
Collapse
Affiliation(s)
- Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Júlia Canet
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Eduard Gracia
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Carme Lluís
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Josefa Mallol
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Enric I. Canela
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Antoni Cortés
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Madrid, Spain
| |
Collapse
|
16
|
Asano T, Takenaga M. Adenosine A 2B Receptors: An Optional Target for the Management of Irritable Bowel Syndrome with Diarrhea? J Clin Med 2017; 6:jcm6110104. [PMID: 29099770 PMCID: PMC5704121 DOI: 10.3390/jcm6110104] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 10/31/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder, with the characteristic symptoms of chronic abdominal pain and altered bowel habits (diarrhea, constipation, or both). IBS is a highly prevalent condition, which negatively affects quality of life and is a significant burden on global healthcare costs. Although many pharmacological medicines have been proposed to treat IBS, including those targeting receptors, channels, and chemical mediators related to visceral hypersensitivity, successful pharmacotherapy for the disease has not been established. Visceral hypersensitivity plays an important role in IBS pathogenesis. Immune activation is observed in diarrhea-predominant patients with IBS and contributes to the development of visceral hypersensitivity. Adenosine is a chemical mediator that regulates many physiological processes, including inflammation and nociception. Among its receptors, the adenosine A2B receptor regulates intestinal secretion, motor function, and the immune response. We recently demonstrated that the adenosine A2B receptor is involved in visceral hypersensitivity in animal models of IBS. In this review, we discuss the possibility of the adenosine A2B receptor as a novel therapeutic target for IBS.
Collapse
Affiliation(s)
- Teita Asano
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki 216-8512, Japan.
| | - Mitsuko Takenaga
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki 216-8512, Japan.
| |
Collapse
|
17
|
Caputi V, Marsilio I, Filpa V, Cerantola S, Orso G, Bistoletti M, Paccagnella N, De Martin S, Montopoli M, Dall'Acqua S, Crema F, Di Gangi I, Galuppini F, Lante I, Bogialli S, Rugge M, Debetto P, Giaroni C, Giron MC. Antibiotic-induced dysbiosis of the microbiota impairs gut neuromuscular function in juvenile mice. Br J Pharmacol 2017; 174:3623-3639. [PMID: 28755521 PMCID: PMC5610159 DOI: 10.1111/bph.13965] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 07/18/2017] [Accepted: 07/19/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice. EXPERIMENTAL APPROACH To induce gut dysbiosis, broad-spectrum antibiotics were administered by gavage to juvenile (3weeks old) male C57Bl/6 mice for 14 days. Bile acid composition in the intestinal lumen was analysed by liquid chromatography-mass spectrometry. Changes in intestinal motility were evaluated by stool frequency, transit of a fluorescent-labelled marker and isometric muscle responses of ileal full-thickness preparations to receptor and non-receptor-mediated stimuli. Alterations in ENS integrity were assessed by immunohistochemistry and Western blot analysis. KEY RESULTS Antibiotic treatment altered gastrointestinal transit, luminal bile acid metabolism and bowel architecture. Gut dysbiosis resulted in distorted glial network, loss of myenteric plexus neurons, altered cholinergic, tachykininergic and nitrergic neurotransmission associated with reduced number of nNOS neurons and different ileal distribution of the toll-like receptor TLR2. Functional defects were partly reversed by activation of TLR2 signalling. CONCLUSIONS AND IMPLICATIONS Gut dysbiosis caused complex morpho-functional neuromuscular rearrangements, characterized by structural defects of the ENS and increased tachykininergic neurotransmission. Altogether, our findings support the beneficial role of enteric microbiota for ENS homeostasis instrumental in ensuring proper gut neuromuscular function during critical stages of development.
Collapse
Affiliation(s)
- Valentina Caputi
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Viviana Filpa
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
- San Camillo HospitalTrevisoItaly
| | - Genny Orso
- IRCCS ‘E. Medea’ Bosisio PariniLeccoItaly
| | | | - Nicola Paccagnella
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Sara De Martin
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of PharmacologyUniversity of PaviaPaviaItaly
| | | | | | | | - Sara Bogialli
- Department of Chemical SciencesUniversity of PadovaPadovaItaly
| | - Massimo Rugge
- Department of MedicineUniversity of PadovaPadovaItaly
| | - Patrizia Debetto
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| | - Cristina Giaroni
- Department of Medicine and SurgeryUniversity of InsubriaVareseItaly
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological SciencesUniversity of PadovaPadovaItaly
| |
Collapse
|
18
|
Pellegrini C, Antonioli L, Colucci R, Tirotta E, Gentile D, Ippolito C, Segnani C, Levandis G, Cerri S, Blandini F, Barocelli E, Ballabeni V, Bernardini N, Blandizzi C, Fornai M. Effects of L-DOPA/benserazide co-treatment on colonic excitatory cholinergic motility and enteric inflammation following dopaminergic nigrostriatal neurodegeneration. Neuropharmacology 2017; 123:22-33. [DOI: 10.1016/j.neuropharm.2017.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/12/2017] [Accepted: 05/15/2017] [Indexed: 01/23/2023]
|
19
|
Antonioli L, Pellegrini C, Fornai M, Tirotta E, Gentile D, Benvenuti L, Giron MC, Caputi V, Marsilio I, Orso G, Bernardini N, Segnani C, Ippolito C, Csóka B, Németh ZH, Haskó G, Scarpignato C, Blandizzi C, Colucci R. Colonic motor dysfunctions in a mouse model of high-fat diet-induced obesity: an involvement of A 2B adenosine receptors. Purinergic Signal 2017; 13:497-510. [PMID: 28808842 DOI: 10.1007/s11302-017-9577-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022] Open
Abstract
Adenosine A2B receptors (A2BR) regulate several enteric functions. However, their implication in the pathophysiology of intestinal dysmotility associated with high-fat diet (HFD)-induced obesity has not been elucidated. We investigated the expression of A2BR in mouse colon and their role in the mechanisms underlying the development of enteric dysmotility associated with obesity. Wild-type C57BL/6J mice were fed with HFD (60% kcal from fat) or normocaloric diet (NCD; 18% kcal from fat) for 8 weeks. Colonic A2BR localization was examined by immunofluorescence. The role of A2BR in the control of colonic motility was examined in functional experiments on longitudinal muscle preparations (LMPs). In NCD mice, A2BR were predominantly located in myenteric neurons; in HFD animals, their expression increased throughout the neuromuscular layer. Functionally, the A2BR antagonist MRS1754 enhanced electrically induced NK1-mediated tachykininergic contractions in LMPs from HFD mice, while it was less effective in tissues from NCD mice. The A2B receptor agonist BAY 60-6583 decreased colonic tachykininergic contractions in LMPs, with higher efficacy in preparations from obese mice. Both A2BR ligands did not affect contractions elicited by exogenous substance P. Obesity is related with a condition of colonic inflammation, leading to an increase of A2BR expression. A2BR, modulating the activity of excitatory tachykininergic nerves, participate to the enteric dysmotility associated with obesity.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.,Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Erika Tirotta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,San Camillo Hospital, Treviso, Italy
| | - Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Genny Orso
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini-, Lecco, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Chiara Ippolito
- 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, Newark, NJ, 07103, USA
| | - Zoltán H Németh
- Department of Surgery, Morristown Medical Center, Morristown, NJ, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | | | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
20
|
Bahreyni A, Samani SS, Khazaei M, Ryzhikov M, Avan A, Hassanian SM. Therapeutic potentials of adenosine receptors agonists and antagonists in colitis; Current status and perspectives. J Cell Physiol 2017; 233:2733-2740. [DOI: 10.1002/jcp.26073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 06/28/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Amirhossein Bahreyni
- Faculty of Medicine; Department of Clinical Biochemistry and Immunogenetic Research Center; Mazandaran University of Medical Sciences; Sari Mazandaran Iran
| | - Seyed S. Samani
- Department of Biology; Mashhad Branch; Islamic Azad University; Mashhad Iran
| | - Majid Khazaei
- Faculty of Medicine; Department of Medical Physiology; Mashhad University of Medical Sciences; Mashhad Iran
| | - Mikhail Ryzhikov
- Department of Molecular Microbiology and Immunology; St. Louis University; School of Medicine; Saint Louis Missouri
| | - Amir Avan
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Department of Modern Sciences and Technologies; School of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Seyed M. Hassanian
- Metabolic Syndrome Research Center; Mashhad University of Medical Sciences; Mashhad Iran
- Faculty of Medicine; Department of Medical Biochemistry; Mashhad University of Medical Sciences; Mashhad Iran
- Microanatomy Research Center; Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
21
|
Zizzo MG, Auteri M, Amato A, Caldara G, Nuzzo D, Di Carlo M, Serio R. Angiotensin II type II receptors and colonic dysmotility in 2,4-dinitrofluorobenzenesulfonic acid-induced colitis in rats. Neurogastroenterol Motil 2017; 29. [PMID: 28160390 DOI: 10.1111/nmo.13019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/10/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Angiotensin II (Ang II), the main peptide of the renin-angiotensin system (RAS), has been suggested to be involved in inflammatory bowel diseases. Since RAS has emerged as gut motility regulator, and dysmotility is associated with intestinal inflammation, our objective was to investigate in rat 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis the functionality of RAS and its contribution to colonic motor alterations. METHODS The effects of Ang II on the longitudinal colonic muscular contractility of control and DNBS-treated rats were characterized in vitro. Transcripts encoding for Ang II receptors were investigated by RT-PCR. KEY RESULTS Inflamed preparations showed a longitudinal muscle marked hypocontractility. Angiotensin II caused contractile effects in both preparations, but the responses in DNBS preparations were reduced compared to controls. In both preparations, Losartan, AT1 receptor antagonist, reduced Ang II effects. PD123319, AT2 receptor antagonist, enhanced Ang II responses only in DNBS rats, as well as Nω -Nitro-L-arginine (L-NNA), nitric oxide (NO) synthase inhibitor, or tetrodotoxin (TTX), neural toxin. The co-administration of PD123319 and TTX or L-NNA produced no additive effects. PD123319 per se improved colonic contractility in inflamed tissues. The effect was reduced in the presence of L-NNA or TTX. All Ang II receptor subtypes were expressed in both preparations. CONCLUSIONS & INFERENCES AT1 receptors mediate Ang II contractile responses in rat colon. During inflammation a recruitment of Ang II AT2 receptors would counteract AT1 -contractile activity. A tonic activation of AT2 receptors would contribute to the general reduction in muscle contractility during experimental inflammation. A role for enteric neurons and NO is also suggested.
Collapse
Affiliation(s)
- M G Zizzo
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Palermo, Italy
| | - M Auteri
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Palermo, Italy
| | - A Amato
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Palermo, Italy
| | - G Caldara
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Palermo, Italy
| | - D Nuzzo
- Istituto di Biomedicina ed Immunologia Molecolare (IBIM) "Alberto Monroy", CNR, Palermo, Italy
| | - M Di Carlo
- Istituto di Biomedicina ed Immunologia Molecolare (IBIM) "Alberto Monroy", CNR, Palermo, Italy
| | - R Serio
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Laboratorio di Fisiologia generale, Università di Palermo, Palermo, Italy
| |
Collapse
|
22
|
Asano T, Tanaka KI, Tada A, Shimamura H, Tanaka R, Maruoka H, Takenaga M, Mizushima T. Aminophylline suppresses stress-induced visceral hypersensitivity and defecation in irritable bowel syndrome. Sci Rep 2017; 7:40214. [PMID: 28054654 PMCID: PMC5214462 DOI: 10.1038/srep40214] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 12/05/2016] [Indexed: 02/08/2023] Open
Abstract
Pharmacological therapy for irritable bowel syndrome (IBS) has not been established. In order to find candidate drugs for IBS with diarrhea (IBS-D), we screened a compound library of drugs clinically used for their ability to prevent stress-induced defecation and visceral hypersensitivity in rats. We selected the bronchodilator aminophylline from this library. Using a specific inhibitor for each subtype of adenosine receptors (ARs) and phosphodiesterases (PDEs), we found that both A2BARs and PDE4 are probably mediated the inhibitory effect of aminophylline on wrap restraint stress (WRS)-induced defecation. Aminophylline suppressed maternal separation- and acetic acid administration-induced visceral hypersensitivity to colorectal distension (CRD), which was mediated by both A2AARs and A2BARs. We propose that aminophylline is a candidate drug for IBS-D because of its efficacy in both of stress-induced defecation and visceral hypersensitivity, as we observed here, and because it is clinically safe.
Collapse
Affiliation(s)
- Teita Asano
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki 216-8512, Japan
| | - Ken-ichiro Tanaka
- Laboratory of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-2αhinmachi, Nishitokyo-shi, 202-8585, Japan
| | - Arisa Tada
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Hikaru Shimamura
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Rikako Tanaka
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Hiroki Maruoka
- Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Mitsuko Takenaga
- Institute of Medical Science, St. Marianna University School of Medicine, 2-16-1, Sugao, Miyamae-ku, Kawasaki 216-8512, Japan
| | - Tohru Mizushima
- LTT Bio-Pharma Co., Ltd, Shiodome Building 3F, 1-2-20 Kaigan, Minato-ku, Tokyo 105-0022, Japan
| |
Collapse
|
23
|
Fornai M, Pellegrini C, Antonioli L, Segnani C, Ippolito C, Barocelli E, Ballabeni V, Vegezzi G, Al Harraq Z, Blandini F, Levandis G, Cerri S, Blandizzi C, Bernardini N, Colucci R. Enteric Dysfunctions in Experimental Parkinson's Disease: Alterations of Excitatory Cholinergic Neurotransmission Regulating Colonic Motility in Rats. J Pharmacol Exp Ther 2016; 356:434-44. [PMID: 26582732 DOI: 10.1124/jpet.115.228510] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/17/2015] [Indexed: 02/06/2023] Open
Abstract
Parkinson's disease is frequently associated with gastrointestinal symptoms, mostly represented by constipation and defecatory dysfunctions. This study examined the impact of central dopaminergic denervation, induced by injection of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle, on distal colonic excitatory cholinergic neuromotor activity in rats. Animals were euthanized 4 and 8 weeks after 6-OHDA injection. In vivo colonic transit was evaluated by radiologic assay. Electrically induced and carbachol-induced cholinergic contractions were recorded in vitro from longitudinal and circular muscle colonic preparations, whereas acetylcholine levels were assayed in the incubation media. Choline acetyltransferase (ChAT), HuC/D (pan-neuronal marker), muscarinic M2 and M3 receptors were assessed by immunohistochemistry or western blot assay. As compared with control rats, at week 4, 6-OHDA-treated animals displayed the following changes: decreased in vivo colonic transit rate, impaired electrically evoked neurogenic cholinergic contractions, enhanced carbachol-induced contractions, decreased basal and electrically stimulated acetylcholine release from colonic tissues, decreased ChAT immunopositivity in the neuromuscular layer, unchanged density of HuC/D immunoreactive myenteric neurons, and increased expression of colonic muscarinic M2 and M3 receptors. The majority of such alterations were also detected at week 8 post 6-OHDA injection. These findings indicate that central nigrostriatal dopaminergic denervation is associated with an impaired excitatory neurotransmission characterized by a loss of myenteric neuronal ChAT positivity and decrease in acetylcholine release, resulting in a dysregulated smooth muscle motor activity, which likely contributes to the concomitant decrease in colonic transit rate.
Collapse
Affiliation(s)
- Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Elisabetta Barocelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Vigilio Ballabeni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Gaia Vegezzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Zainab Al Harraq
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Fabio Blandini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Giovanna Levandis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Silvia Cerri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| | - Rocchina Colucci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy (M.F., C.P., L.A., C.S., C.I., C.B., N.B.); Department of Pharmacy, University of Parma, Parma, Italy (E.B., V.B., G.V., Z.A.H.); Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurologic Institute, Pavia, Italy (F.B., G.L., S.C.); and Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy (R.C.)
| |
Collapse
|
24
|
Gonçalves FQ, Pires J, Pliassova A, Beleza R, Lemos C, Marques JM, Rodrigues RJ, Canas PM, Köfalvi A, Cunha RA, Rial D. Adenosine A2b receptors control A1 receptor-mediated inhibition of synaptic transmission in the mouse hippocampus. Eur J Neurosci 2015; 41:878-88. [PMID: 25704806 DOI: 10.1111/ejn.12851] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/29/2014] [Accepted: 01/14/2015] [Indexed: 01/21/2023]
Abstract
Adenosine is a neuromodulator mostly acting through A1 (inhibitory) and A2A (excitatory) receptors in the brain. A2B receptors (A(2B)R) are G(s/q)--protein-coupled receptors with low expression in the brain. As A(2B)R function is largely unknown, we have now explored their role in the mouse hippocampus. We performed electrophysiological extracellular recordings in mouse hippocampal slices, and immunological analysis of nerve terminals and glutamate release in hippocampal slices and synaptosomes. Additionally, A(2B)R-knockout (A(2B)R-KO) and C57/BL6 mice were submitted to a behavioural test battery (open field, elevated plus-maze, Y-maze). The A(2B)R agonist BAY60-6583 (300 nM) decreased the paired-pulse stimulation ratio, an effect prevented by the A(2B)R antagonist MRS 1754 (200 nM) and abrogated in A(2B)R-KO mice. Accordingly, A(2B)R immunoreactivity was present in 73 ± 5% of glutamatergic nerve terminals, i.e. those immunopositive for vesicular glutamate transporters. Furthermore, BAY 60-6583 attenuated the A(1)R control of synaptic transmission, both the A(1)R inhibition caused by 2-chloroadenosine (0.1-1 μM) and the disinhibition caused by the A(1)R antagonist DPCPX (100 nM), both effects prevented by MRS 1754 and abrogated in A(2B)R-KO mice. BAY 60-6583 decreased glutamate release in slices and also attenuated the A(1)R inhibition (CPA 100 nM). A(2B)R-KO mice displayed a modified exploratory behaviour with an increased time in the central areas of the open field, elevated plus-maze and the Y-maze and no alteration of locomotion, anxiety or working memory. We conclude that A(2B)R are present in hippocampal glutamatergic terminals where they counteract the predominant A(1)R-mediated inhibition of synaptic transmission, impacting on exploratory behaviour.
Collapse
Affiliation(s)
- Francisco Q Gonçalves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517, Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Antonioli L, Giron MC, Colucci R, Pellegrini C, Sacco D, Caputi V, Orso G, Tuccori M, Scarpignato C, Blandizzi C, Fornai M. Involvement of the P2X7 purinergic receptor in colonic motor dysfunction associated with bowel inflammation in rats. PLoS One 2014; 9:e116253. [PMID: 25549098 PMCID: PMC4280204 DOI: 10.1371/journal.pone.0116253] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 12/06/2014] [Indexed: 12/31/2022] Open
Abstract
Background and Purpose Recent evidence indicates an involvement of P2X7 purinergic receptor (P2X7R) in the fine tuning of immune functions, as well as in driving enteric neuron apoptosis under intestinal inflammation. However, the participation of this receptor in the regulation of enteric neuromuscular functions remains undetermined. This study was aimed at investigating the role of P2X7Rs in the control of colonic motility in experimental colitis. Experimental Approach Colitis was induced in rats by 2,4-dinitrobenzenesulfonic acid. P2X7R distribution was examined by immunofluorescence analysis. The effects of A804598 (selective P2X7R antagonist) and BzATP (P2X7R agonist) were tested on contractions of longitudinal smooth muscle evoked by electrical stimulation or by carbachol in the presence of tetrodotoxin. Key Results P2X7Rs were predominantly located in myenteric neurons, but, in the presence of colitis, their expression increased in the neuromuscular layer. In normal preparations, A804598 elicited a negligible increase in electrically induced contractions, while a significant enhancement was recorded in inflamed tissues. In the presence of Nω-propyl-L-arginine (NPA, neuronal nitric oxide synthase inhibitor) the A804598 effects were lost. P2X7R stimulation with BzATP did not significantly affect electrical-induced contractions in normal colon, while a marked reduction was recorded under inflammation. The inhibitory effect of BzATP was antagonized by A804598, and it was also markedly blunted by NPA. Both P2X7R ligands did not affect carbachol-induced contractions. Conclusions and Implications The purinergic system contributes to functional neuromuscular changes associated with bowel inflammation via P2X7Rs, which modulate the activity of excitatory cholinergic nerves through a facilitatory control on inhibitory nitrergic pathways.
Collapse
Affiliation(s)
- Luca Antonioli
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Rocchina Colucci
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- * E-mail:
| | - Carolina Pellegrini
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Deborah Sacco
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Genny Orso
- Scientific Institute IRCCS Eugenio Medea, Conegliano, Treviso, Italy
| | - Marco Tuccori
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Corrado Blandizzi
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Division of Pharmacology and Chemotherapy, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
The role of extracellular ATP-mediated purinergic signaling in bone, cartilage, and tooth tissue. J Oral Biosci 2014. [DOI: 10.1016/j.job.2014.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
27
|
Feed-forward inhibition of CD73 and upregulation of adenosine deaminase contribute to the loss of adenosine neuromodulation in postinflammatory ileitis. Mediators Inflamm 2014; 2014:254640. [PMID: 25210228 PMCID: PMC4152956 DOI: 10.1155/2014/254640] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 01/31/2023] Open
Abstract
Purinergic signalling is remarkably plastic during gastrointestinal inflammation. Thus, selective drugs targeting the “purinome” may be helpful for inflammatory gastrointestinal diseases. The myenteric neuromuscular transmission of healthy individuals is fine-tuned and controlled by adenosine acting on A2A excitatory receptors. Here, we investigated the neuromodulatory role of adenosine in TNBS-inflamed longitudinal muscle-myenteric plexus of the rat ileum. Seven-day postinflammation ileitis lacks adenosine neuromodulation, which may contribute to acceleration of gastrointestinal transit. The loss of adenosine neuromodulation results from deficient accumulation of the nucleoside at the myenteric synapse despite the fact that the increases in ATP release were observed. Disparity between ATP outflow and adenosine deficit in postinflammatory ileitis is ascribed to feed-forward inhibition of ecto-5′-nucleotidase/CD73 by high extracellular ATP and/or ADP. Redistribution of NTPDase2, but not of NTPDase3, from ganglion cell bodies to myenteric nerve terminals leads to preferential ADP accumulation from released ATP, thus contributing to the prolonged inhibition of muscle-bound ecto-5′-nucleotidase/CD73 and to the delay of adenosine formation at the inflamed neuromuscular synapse. On the other hand, depression of endogenous adenosine accumulation may also occur due to enhancement of adenosine deaminase activity. Both membrane-bound and soluble forms of ecto-5′-nucleotidase/CD73 and adenosine deaminase were detected in the inflamed myenteric plexus. These findings provide novel therapeutic targets for inflammatory gut motility disorders.
Collapse
|