1
|
Karagianni E, Rassouli O, Poulaki S, Dermitzaki E, Liapakis G, Margioris AN, Venihaki M. Corticotropin-releasing hormone deficiency results in impaired analgesic response during CFA-induced inflammation. Hormones (Athens) 2024; 23:535-545. [PMID: 38740711 PMCID: PMC11436445 DOI: 10.1007/s42000-024-00565-8] [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: 11/10/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024]
Abstract
PURPOSE Corticotropin-releasing hormone (CRH) plays an important role in relief of pain by releasing analgesia-associated molecules in several inflammatory states. During inflammation, peripheral CRH acts on cells of the immune system to stimulate the local expression of proopiomelanocortin (POMC) and the production of β-endorphin, which in turn binds to opioid receptors on sensory neurons to produce antinociception. In the present study, we further investigated the role of endogenous CRH in inflammatory pain by determining the effects of Crh-deficiency on this process. METHODS For this purpose, we used Crh-deficient (Crh-/-) mice and their wildtype (Crh + / +) littermates in the CFA (Complete Freund's Adjuvant)-induced inflammatory pain model. Pain thresholds were evaluated with the Hargreaves apparatus. RESULTS Our experiments showed that Crh deficiency led to increased pain response, which was associated with decreased POMC mRNA levels in locally inflamed paws of these mice. Furthermore, Crh-/- mice had higher paw edema than Crh + / + mice. Histological evaluation of inflamed paw tissues revealed increased inflammatory response in Crh-/- mice. Protein levels of proinflammatory cytokines, such as IL-6, TNF-α, and IL-1β, were higher in inflamed tissue of Crh-/- mice compared to wildtype mice. Corticosterone replacement increased the pain threshold of Crh-/- mice, restored their paw volume to the levels of wildtype mice, and significantly reduced their proinflammatory cytokine levels. Furthermore, glucocorticoid administration significantly increased POMC mRNA expression in the inflamed paw. CONCLUSION Our data suggest that genetic deficiency of CRH is associated with increased pain. This effect is likely attributable to the accompanying glucocorticoid insufficiency and is in part mediated by opioids expressed locally.
Collapse
Affiliation(s)
- Efthymia Karagianni
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Olga Rassouli
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Smaragda Poulaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Eirini Dermitzaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - George Liapakis
- Department of Pharmacology, School of Medicine, University of Crete, Heraklion, Greece
| | - Andrew N Margioris
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece
| | - Maria Venihaki
- Department of Clinical Chemistry, School of Medicine, University of Crete, Heraklion, Greece.
| |
Collapse
|
2
|
Swierczynski M, Kasprzak Z, Makaro A, Salaga M. Regulators of G-Protein Signaling (RGS) in Sporadic and Colitis-Associated Colorectal Cancer. Int J Mol Sci 2024; 25:577. [PMID: 38203748 PMCID: PMC10778579 DOI: 10.3390/ijms25010577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/21/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common neoplasms worldwide. Among the risk factors of CRC, inflammatory bowel disease (IBD) is one of the most important ones leading to the development of colitis-associated CRC (CAC). G-protein coupled receptors (GPCR) are transmembrane receptors that orchestrate a multitude of signaling cascades in response to external stimuli. Because of their functionality, they are promising targets in research on new strategies for CRC diagnostics and treatment. Recently, regulators of G-proteins (RGS) have been attracting attention in the field of oncology. Typically, they serve as negative regulators of GPCR responses to both physiological stimuli and medications. RGS activity can lead to both beneficial and harmful effects depending on the nature of the stimulus. However, the atypical RGS-AXIN uses its RGS domain to antagonize key signaling pathways in CRC development through the stabilization of the β-catenin destruction complex. Since AXIN does not limit the efficiency of medications, it seems to be an even more promising pharmacological target in CRC treatment. In this review, we discuss the current state of knowledge on RGS significance in sporadic CRC and CAC with particular emphasis on the regulation of GPCR involved in IBD-related inflammation comprising opioid, cannabinoid and serotonin receptors.
Collapse
Affiliation(s)
| | | | | | - Maciej Salaga
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (M.S.); (Z.K.); (A.M.)
| |
Collapse
|
3
|
Mas-Orea X, Rey L, Battut L, Bories C, Petitfils C, Abot A, Gheziel N, Wemelle E, Blanpied C, Motta JP, Knauf C, Barreau F, Espinosa E, Aloulou M, Cenac N, Serino M, Mouledous L, Fazilleau N, Dietrich G. Proenkephalin deletion in hematopoietic cells induces intestinal barrier failure resulting in clinical feature similarities with irritable bowel syndrome in mice. Commun Biol 2023; 6:1168. [PMID: 37968381 PMCID: PMC10652007 DOI: 10.1038/s42003-023-05542-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/03/2023] [Indexed: 11/17/2023] Open
Abstract
Opioid-dependent immune-mediated analgesic effects have been broadly reported upon inflammation. In preclinical mouse models of intestinal inflammatory diseases, the local release of enkephalins (endogenous opioids) by colitogenic T lymphocytes alleviate inflammation-induced pain by down-modulating gut-innervating nociceptor activation in periphery. In this study, we wondered whether this immune cell-derived enkephalin-mediated regulation of the nociceptor activity also operates under steady state conditions. Here, we show that chimeric mice engrafted with enkephalin-deficient bone marrow cells exhibit not only visceral hypersensitivity but also an increase in both epithelial paracellular and transcellular permeability, an alteration of the microbial topography resulting in increased bacteria-epithelium interactions and a higher frequency of IgA-producing plasma cells in Peyer's patches. All these alterations of the intestinal homeostasis are associated with an anxiety-like behavior despite the absence of an overt inflammation as observed in patients with irritable bowel syndrome. Thus, our results show that immune cell-derived enkephalins play a pivotal role in maintaining gut homeostasis and normal behavior in mice. Because a defect in the mucosal opioid system remarkably mimics some major clinical symptoms of the irritable bowel syndrome, its identification might help to stratify subgroups of patients.
Collapse
Affiliation(s)
- Xavier Mas-Orea
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Lea Rey
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Louise Battut
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Cyrielle Bories
- INFINITy, Université de Toulouse, INSERM U1291, CNRS U5051, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Camille Petitfils
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Anne Abot
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
- Enterosys SAS, Labège, France
| | - Nadine Gheziel
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
- INFINITy, Université de Toulouse, INSERM U1291, CNRS U5051, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Eve Wemelle
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Catherine Blanpied
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Jean-Paul Motta
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Claude Knauf
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Frederick Barreau
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Eric Espinosa
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Meryem Aloulou
- INFINITy, Université de Toulouse, INSERM U1291, CNRS U5051, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Nicolas Cenac
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Matteo Serino
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Lionel Mouledous
- Research Center on Animal Cognition (CRCA), Center of Integrative Biology (CBI), Université de Toulouse, CNRS UMR-5169, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Nicolas Fazilleau
- INFINITy, Université de Toulouse, INSERM U1291, CNRS U5051, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Gilles Dietrich
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
4
|
Sandoval-Caballero C, Luarte L, Jiménez Y, Jaque C, Cifuentes F, Arenas GA, Figueroa M, Jara J, Olszewski PK, Teske JA, Pérez-Leighton CE. Meta-analysis of pre-clinical studies on the effects of opioid receptor ligands on food intake, motivation, and choice. Neurosci Biobehav Rev 2023; 152:105288. [PMID: 37331611 DOI: 10.1016/j.neubiorev.2023.105288] [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: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
The opioid receptors (OR) regulate food intake. Still, despite extensive pre-clinical research, the overall effects and individual contribution of the mu (MOR), kappa (KOR), and delta (DOR) OR subtypes to feeding behaviors and food intake remain unclear. To address this, we conducted a pre-registered systematic search and meta-analysis of rodent dose-response studies to evaluate the impact of central and peripheral administration of non-selective and selective OR ligands on intake, motivation, and choice of food. All studies had a high bias risk. Still, the meta-analysis confirmed the overall orexigenic and anorexigenic effects of OR agonists and antagonists, respectively. Our results support a larger orexigenic role for central MOR agonists among OR subtypes and that peripheral OR antagonists reduce motivation for and intake of preferred foods. In binary food choice studies, peripheral OR agonists selectively increase the intake of fat-preferred foods; in contrast, they did not increase the intake of sweet carbohydrate-preferred foods. Overall, these data support that OR regulation of intake, motivation, and choice is influenced by food macronutrient composition.
Collapse
Affiliation(s)
- C Sandoval-Caballero
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - L Luarte
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - Y Jiménez
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - C Jaque
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - F Cifuentes
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - G A Arenas
- Instituto de Ciencias de la Ingeniería, Universidad de O'Higgins, Libertador Bernardo O'Higgins #611, Rancagua 2841959, Region del Libertador Bernardo O'Higgins, Chile
| | - M Figueroa
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - J Jara
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile
| | - P K Olszewski
- Faculty of Science and Engineering, University of Waikato, Hamilton, Private Bag 3105, Hamilton 3240, New Zealand
| | - J A Teske
- School of Nutritional Sciences and Wellness and the Graduate Interdisciplinary Programs in Physiological Sciences and Neuroscience at the University of Arizona, 1177 E 4th Street Shantz 332, Tucson, AZ 85721, USA
| | - C E Pérez-Leighton
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Avenida Libertador Bernardo O'Higgins 340, Santiago 8331150, Region Metropolitana, Chile.
| |
Collapse
|
5
|
Manai F, Zanoletti L, Morra G, Mansoor S, Carriero F, Bozzola E, Muscianisi S, Comincini S. Gluten Exorphins Promote Cell Proliferation through the Activation of Mitogenic and Pro-Survival Pathways. Int J Mol Sci 2023; 24:3912. [PMID: 36835317 PMCID: PMC9966116 DOI: 10.3390/ijms24043912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Celiac disease (CD) is a chronic and systemic autoimmune disorder that affects preferentially the small intestine of individuals with a genetic predisposition. CD is promoted by the ingestion of gluten, a storage protein contained in the endosperm of the seeds of wheat, barley, rye, and related cereals. Once in the gastrointestinal (GI) tract, gluten is enzymatically digested with the consequent release of immunomodulatory and cytotoxic peptides, i.e., 33mer and p31-43. In the late 1970s a new group of biologically active peptides, called gluten exorphins (GEs), was discovered and characterized. In particular, these short peptides showed a morphine-like activity and high affinity for the δ-opioid receptor (DOR). The relevance of GEs in the pathogenesis of CD is still unknown. Recently, it has been proposed that GEs could contribute to asymptomatic CD, which is characterized by the absence of symptoms that are typical of this disorder. In the present work, GEs cellular and molecular effects were in vitro investigated in SUP-T1 and Caco-2 cells, also comparing viability effects with human normal primary lymphocytes. As a result, GEs treatments increased tumor cell proliferation by cell cycle and Cyclins activation as well as by induction of mitogenic and pro-survival pathways. Finally, a computational model of GEs interaction with DOR is provided. Altogether, the results might suggest a possible role of GEs in CD pathogenesis and on its associated cancer comorbidities.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
- Laboratory for Mucosal Immunology, TARGID, Department of Chronic Diseases, Metabolism and Ageing, KU Leuven, 3000 Leuven, Belgium
| | - Giulia Morra
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Samman Mansoor
- SCITEC, Consiglio Nazionale delle Ricerche, 20131 Milano, Italy
| | - Francesca Carriero
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| | - Elena Bozzola
- Pediatric Unit, I.R.C.C.S. Bambino Gesù Children Hospital, 00165 Roma, Italy
| | - Stella Muscianisi
- Cell Factory and Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology “L.Spallanzani”, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
6
|
Pang Y, Zheng Y, Yang N, Zan M, Zhang L, Ding W. Potential novel biomarkers in small intestine for obesity/obesity resistance revealed by multi-omics analysis. Lipids Health Dis 2022; 21:98. [PMID: 36209126 PMCID: PMC9547412 DOI: 10.1186/s12944-022-01711-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Background Although obesity is caused by different factors, individual susceptibility to obesity differs among people under the same circumstances. The microbiota in the caecum or fresh faeces and metabolites in blood or urine contribute to obesity resistance; however, the microbiota or metabolites in the small intestine have not been extensively studied. Methods To investigate the relationship between the microbiota or metabolites in the small intestine and susceptibility to obesity, eighty-eight male C57BL/6 mice were fed a high-fat diet (HFD) for 8 weeks to establish two models of obesity and obesity resistance. For further study, six mice were chosen from among the obesity models, and twelve mice were randomly chosen from among the obesity resistance models. After fasting plasma glucose and behavioural testing, the mice were fed in single cages for another 4 weeks to observe their weight and food intake. All mice were sacrificed at 20 weeks of age. Serum ALT, AST, HDL, LDL, TG and TC levels were measured using an automatic biochemical analyser. The microbiota and metabolites in the small intestine contents were analysed using 16 S sequencing and an ultrahigh-performance liquid chromatographic system, respectively. Transcripts in the jejunum were evaluated using full-length transcriptome sequencing and verified by qPCR. Results The results showed that HFD induced depression and anxiety behaviours and higher fasting plasma glucose, ALT, AST, HDL, LDL, TG and TC levels in the obese mice; however, these levels were improved in obese resistance mice. The correlation analysis showed that the phosphatidylcholine, TG, and phosphatidylethanolamine levels were higher in obese mice and correlated positively with intestinal microflora (Desulfovibrio and Gemella) and the Cxcl10 gene. A higher abundance of Clostridium_sensu_stricto_1 in obesity-resistant mice correlated negatively with the metabolite contents (neuromedin N and enkephalin L) and Pck1 gene expression and correlated positively with certain metabolites (5-hydroxy-L-tryptophan, cinnamyl alcohol and 1 H-indole-3-acetamide) and genes expression (Gdf15, Igfbp6 and Spp1). Conclusion Clostridium_sensu_stricto_1, neuromedin N, enkephalin L, Pck1, 5-hydroxy-L-tryptophan, Cxcl10 and cinnamyl alcohol may be novel biomarkers in the small intestine for obesity/obesity resistance. These might be helpful for obesity prevention or for treating obese patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-022-01711-0.
Collapse
Affiliation(s)
- Yueshan Pang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.,The Second Clinical Medical College, North SiChuan Medical College, 637000, Nanchong, China
| | - Yali Zheng
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Ni Yang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Meng Zan
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - Lu Zhang
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China
| | - WeiJun Ding
- Department of Fundamental Medicine, Chengdu University of Traditional Chinese Medicine, 611130, Chengdu, China.
| |
Collapse
|
7
|
The role of kappa opioid receptors in immune system - An overview. Eur J Pharmacol 2022; 933:175214. [PMID: 36007608 DOI: 10.1016/j.ejphar.2022.175214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 11/20/2022]
Abstract
Opioids are one of the most effective anti-nociceptive agents used in patients with cancer pain or after serious surgery in most countries. The endogenous opioid system participates in pain perception, but recently its role in inflammation was determined. κ-opioid receptors (KOP receptors), a member of the opioid receptor family, are expressed in the central and peripheral nervous system as well as on the surface of different types of immune cells, e.g. T cells, B cells and monocytes. In this review, we focused on the involvement of KOP receptors in the inflammatory process and described their function in a number of conditions in which the immune system plays a key role (e.g. inflammatory bowel disease, arthritis, subarachnoid hemorrhage, vascular dysfunction) and inflammatory pain. We summed up the application of known KOP ligands in pathophysiology and we aimed to shed new light on KOP receptors as important elements during inflammation.
Collapse
|
8
|
Mas-Orea X, Basso L, Blanpied C, Gaveriaux-Ruff C, Cenac N, Dietrich G. Delta opioid receptors on nociceptive sensory neurons mediate peripheral endogenous analgesia in colitis. J Neuroinflammation 2022; 19:7. [PMID: 34991641 PMCID: PMC8740424 DOI: 10.1186/s12974-021-02352-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Inflammatory visceral pain is endogenously controlled by enkephalins locally released by mucosal CD4+ T lymphocytes in mice. The present study aimed at identifying opioid receptor(s) expressed on nociceptive sensory nerves involved in this peripheral opioid-mediated analgesia. METHODS The peripheral analgesia associated with the accumulation of CD4+ T lymphocytes within the inflamed colonic mucosa was assessed in conditional knockout mice specifically deleted for either of the two opioid receptors for enkephalins (i.e., µ (MOR) and δ (DOR) receptors) in Nav1.8-expressing sensory neurons in the dextran sulfate sodium (DSS)-induced colitis model. RESULTS Endogenous analgesia is lost in conditional knockout mice for DOR, but not MOR at the later phase of the DSS-induced colitis. The absence of either of the opioid receptors on sensory nerves had no impact on both the colitis severity and the rate of T lymphocytes infiltrating the inflamed colonic mucosa. CONCLUSION The key role of DOR on primary afferents in relieving intestinal inflammatory pain opens new therapeutic opportunities for peripherally restricted DOR analgesics to avoid most of the side effects associated with MOR-targeting drugs used in intestinal disorders.
Collapse
Affiliation(s)
- Xavier Mas-Orea
- Digestive Health Research Institute (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan BP 3028, 31024, Toulouse Cedex 3, France
| | - Lilian Basso
- Digestive Health Research Institute (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan BP 3028, 31024, Toulouse Cedex 3, France
- INFINITy, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Catherine Blanpied
- Digestive Health Research Institute (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan BP 3028, 31024, Toulouse Cedex 3, France
| | | | - Nicolas Cenac
- Digestive Health Research Institute (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan BP 3028, 31024, Toulouse Cedex 3, France
| | - Gilles Dietrich
- Digestive Health Research Institute (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, CHU Purpan BP 3028, 31024, Toulouse Cedex 3, France.
| |
Collapse
|
9
|
Bobo TR, Fitzpatrick LR, Whitcomb TL, Cooper TK, Raiciulescu S, Smith JP. Role of the δ-Opioid Receptor in 2 Murine Models of Colitis. Comp Med 2020; 70:25-34. [PMID: 31969211 DOI: 10.30802/aalas-cm-19-000024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Crohn disease and ulcerative colitis, collectively referred to as inflammatory bowel disease (IBD), are chronic inflammatory disorders of the gastrointestinal tract. Currently, the etiology of IBD is unknown, and immunosuppressive therapies have become the standard of care to reduce the inflammation; however, these agents only induce remission 50% of the time in patients and can have serious side effects. Recently, endogenous opioids and opioid receptors have been shown to play a role in the mediation of inflammation. In addition, opioid receptor blockade with a nonselective antagonist, naltrexone, has been shown to reduce colitis in both murine models and human subjects. The goal of the current study was to determine if the antiinflammatory effects of naltrexone are mediated through the delta (δ) opioid receptor. Male C57BL/6NCrl (6 to 8 wk.; n = 110) and female BALB/cAnNCrl (6-8 wk.; n = 91) mice were studied using 2 animal models of chemically induced colitis: dextran sodium sulfate (DSS) and 2, 4, 6-trinitrobenzenesulfonic acid (TNBS). The selective δ-receptor antagonists naltrindole and 7-benzylidenenaltrexone were administered to examine the role of the δ-opioid receptor in colonic inflammation. The quantitative measurement of colitis activity, colon weight and length, Hct, WBC count, and gross and microscopic aberrations were analyzed. Administration of naltrexone in the DSS colitis model significantly improved overall disease activity indices on day 5 of therapy. The use of δ-antagonists and naltrexone had limited to no effect on TNBS colitis. Similar findings were obtained by using the DSS colitis model. Based on the current findings, the authors conclude that naltrexone therapy has limited effect on the improvement of colitis in 2 murine models; however, the δ-opioid receptor was not responsible for mediating the effects.
Collapse
Affiliation(s)
- Tia R Bobo
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, Pennsylvania; Office of Animal Research, The George Washington University, Washington DC;,
| | - Leo R Fitzpatrick
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania; Department of Pharmaceutical & Biomedical Sciences, California Northstate University, Elk Grove, California
| | - Tiffany L Whitcomb
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, Pennsylvania
| | - Timothy K Cooper
- Department of Comparative Medicine, Penn State College of Medicine, Hershey, Pennsylvania; Department of Pathology, Penn State College of Medicine, Hershey, Pennsylvania; Charles River Laboratories, Contractor Supporting National Institute of Allergy and Infectious Disease, Frederick, Maryland
| | - Sorana Raiciulescu
- Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, Maryland
| | - Jill P Smith
- Department of Medicine, Penn State College of Medicine, Hershey, Pennsylvania; Department of Medicine, Georgetown University, Washington, DC
| |
Collapse
|
10
|
Gupta K, Chen C, Lutty GA, Hebbel RP. Morphine promotes neovascularizing retinopathy in sickle transgeneic mice. Blood Adv 2019; 3:1073-1083. [PMID: 30944099 PMCID: PMC6457224 DOI: 10.1182/bloodadvances.2018026898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/28/2019] [Indexed: 12/11/2022] Open
Abstract
Neovascularizing retinopathy is a significant complication of sickle cell disease (SCD), occurring more frequently in HbSC than HbSS disease. This risk difference is concordant with a divergence of angiogenesis risk, as identified by levels of pro- vs anti-angiogenic factors in the sickle patient's blood. Because our prior studies documented that morphine promotes angiogenesis in both malignancy and wound healing, we tested whether chronic opioid treatment would promote retinopathy in NY1DD sickle transgenic mice. After 10 to 15 months of treatment, sickle mice treated with morphine developed neovascularizing retinopathy to a far greater extent than either of the controls (sickle mice treated with saline and wild-type mice treated identically with morphine). Our dissection of the mechanistic linkage between morphine and retinopathy revealed a complex interplay among morphine engagement with its μ opioid receptor (MOR) on retinal endothelial cells (RECs); morphine-induced production of tumor necrosis factor α and interleukin-6 (IL-6), causing increased expression of both MOR and vascular endothelial growth factor receptor 2 (VEGFR2) on RECs; morphine/MOR engagement transactivating VEGFR2; and convergence of MOR, VEGFR2, and IL-6 activation on JAK/STAT3-dependent REC proliferation and angiogenesis. In the NY1DD mice, the result was increased angiogenesis, seen as neovascularizing retinopathy, similar to the retinal pathology occurring in humans with SCD. Therefore, we conclude that chronic opioid exposure, superimposed on the already angiogenic sickle milieu, might enhance risk for retinopathy. These results provide an additional reason for development and application of opioid alternatives for pain control in SCD.
Collapse
Affiliation(s)
- Kalpna Gupta
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| | - Chunsheng Chen
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| | - Gerard A Lutty
- Wilmer Ophthalmological Institute, John Hopkins School of Medicine, Baltimore, MD
| | - Robert P Hebbel
- Vascular Biology Center, and
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN; and
| |
Collapse
|
11
|
DiCello JJ, Saito A, Rajasekhar P, Eriksson EM, McQuade RM, Nowell CJ, Sebastian BW, Fichna J, Veldhuis NA, Canals M, Bunnett NW, Carbone SE, Poole DP. Inflammation-associated changes in DOR expression and function in the mouse colon. Am J Physiol Gastrointest Liver Physiol 2018; 315:G544-G559. [PMID: 29927325 PMCID: PMC6230691 DOI: 10.1152/ajpgi.00025.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Endogenous opioids activate opioid receptors (ORs) in the enteric nervous system to control intestinal motility and secretion. The μ-OR mediates the deleterious side effects of opioid analgesics, including constipation, respiratory depression, and addiction. Although the δ-OR (DOR) is a promising target for analgesia, the function and regulation of DOR in the colon are poorly understood. This study provides evidence that endogenous opioids activate DOR in myenteric neurons that may regulate colonic motility. The DOR agonists DADLE, deltorphin II, and SNC80 inhibited electrically evoked contractions and induced neurogenic contractions in the mouse colon. Electrical, chemical, and mechanical stimulation of the colon evoked the release of endogenous opioids, which stimulated endocytosis of DOR in the soma and proximal neurites of myenteric neurons of transgenic mice expressing DOR fused to enhanced green fluorescent protein. In contrast, DOR was not internalized in nerve fibers within the circular muscle. Administration of dextran sulfate sodium induced acute colitis, which was accompanied by DOR endocytosis and an increased density of DOR-positive nerve fibers within the circular muscle. The potency with which SNC80 inhibited neurogenic contractions was significantly enhanced in the inflamed colon. This study demonstrates that DOR-expressing neurons in the mouse colon can be activated by exogenous and endogenous opioids. Activated DOR traffics to endosomes and inhibits neurogenic motility of the colon. DOR signaling is enhanced during intestinal inflammation. This study demonstrates functional expression of DOR by myenteric neurons and supports the therapeutic targeting of DOR in the enteric nervous system. NEW & NOTEWORTHY DOR is activated during physiologically relevant reflex stimulation. Agonist-evoked DOR endocytosis is spatially and temporally regulated. A significant proportion of DOR is internalized in myenteric neurons during inflammation. The relative proportion of all myenteric neurons that expressed DOR and the overlap with the nNOS-positive population are increased in inflammation. DOR-specific innervation of the circular muscle is increased in inflammation, and this is consistent with enhanced responsiveness to the DOR agonist SNC80.
Collapse
Affiliation(s)
- Jesse J. DiCello
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Ayame Saito
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Pradeep Rajasekhar
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Emily M. Eriksson
- 2Division of Population Health and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia,3Division of Infection and Immunity, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia,4Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Rachel M. McQuade
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Cameron J. Nowell
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Benjamin W. Sebastian
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jakub Fichna
- 5Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Nicholas A. Veldhuis
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,6Department of Genetics, University of Melbourne, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Meritxell Canals
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Nigel W. Bunnett
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia,8Department of Pharmacology and Therapeutics University of Melbourne, Parkville, Victoria, Australia,9Department of Surgery and Pharmacology, Columbia University, New York, New York
| | - Simona E. Carbone
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia
| | - Daniel P. Poole
- 1Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia,7ARC Centre of Excellence in Bio-Nano Science and Technology, Parkville, Victoria, Australia,10Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
The influence of experimental inflammation and axotomy on leucine enkephalin (leuENK) distribution in intramural nervous structures of the porcine descending colon. BMC Vet Res 2018; 14:169. [PMID: 29793486 PMCID: PMC5968568 DOI: 10.1186/s12917-018-1496-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 05/14/2018] [Indexed: 12/13/2022] Open
Abstract
Background The enteric nervous system (ENS), located in the intestinal wall and characterized by considerable independence from the central nervous system, consists of millions of cells. Enteric neurons control the majority of functions of the gastrointestinal tract using a wide range of substances, which are neuromediators and/or neuromodulators. One of them is leucine–enkephalin (leuENK), which belongs to the endogenous opioid family. It is known that opioids in the gastrointestinal tract have various functions, including visceral pain conduction, intestinal motility and secretion and immune processes, but many aspects of distribution and function of leuENK in the ENS, especially during pathological states, remain unknown. Results During this experiment, the distribution of leuENK – like immunoreactive (leuENK-LI) nervous structures using the immunofluorescence technique were studied in the porcine colon in physiological conditions, during chemically-induced inflammation and after axotomy. The study included the circular muscle layer, myenteric (MP), outer submucous (OSP) and inner submucous plexus (ISP) and the mucosal layer. In control animals, the number of leuENK-LI neurons amounted to 4.86 ± 0.17%, 2.86 ± 0.28% and 1.07 ± 0.08% in the MP, OSP and ISP, respectively. Generally, both pathological stimuli caused an increase in the number of detected leuENK-LI cells, but the intensity of the observed changes depended on the factor studied and part of the ENS. The percentage of leuENK-LI perikarya amounted to 11.48 ± 0.96%, 8.71 ± 0.13% and 9.40 ± 0.76% during colitis, and 6.90 ± 0.52% 8.46 ± 12% and 4.48 ± 0.44% after axotomy in MP, OSP and ISP, respectively. Both processes also resulted in an increase in the number of leuENK-LI nerves in the circular muscle layer, whereas changes were less visible in the mucosa during inflammation and axotomy did not change the number of leuENK-LI mucosal fibers. Conclusions LeuENK in the ENS takes part in intestinal regulatory processes not only in physiological conditions, but also under pathological factors. The observed changes are probably connected with the participation of leuENK in sensory and motor innervation and the neuroprotective effects of this substance. Differences in the number of leuENK-LI neurons during inflammation and after axotomy may suggest that the exact functions of leuENK probably depend on the type of pathological factor acting on the intestine.
Collapse
|
13
|
Wojtkiewicz J, Rytel L, Makowska K, Gonkowski S. Co-localization of zinc transporter 3 (ZnT3) with sensory neuromediators and/or neuromodulators in the enteric nervous system of the porcine esophagus. Biometals 2017; 30:393-403. [PMID: 28417221 PMCID: PMC5425499 DOI: 10.1007/s10534-017-0014-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/29/2017] [Indexed: 01/27/2023]
Abstract
Zinc transporter 3 (ZnT3) is one of the zinc transporters family. It is closely connected to the nervous system, where enables the transport of zinc ions from the cytoplasm to synaptic vesicles. This substance has been described within the central and peripheral nervous system, especially in the enteric nervous system (ENS). The aim of the present study was to describe the co-localization of ZnT3 with selected neuromediators and/or neuromodulators participating in sensory stimuli conduction in neurons of the ENS within the porcine esophagus. Co-localization of ZnT3 with substance P (SP), leucine enkephalin (LENK) and calcitonin gene-related peptide (CGRP) was studied using standard double-immunofluorescence technique. The obtained results show that ZnT3, SP and/or LENK may occur in the same enteric neurons, and the degree of co-localization of these substances clearly depends on the fragment of esophagus studied and the type of enteric ganglia. In contrast, the co-localization of ZnT3 with CGRP was not observed during the present investigation. The obtained results suggest that ZnT3 in the ENS may be involved in the conduction of sensory and/or pain stimuli.
Collapse
Affiliation(s)
- Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland. .,Laboratory for Regenerative Medicine, Faculty of Medical Sciences, University of Warmia and Mazury, Olsztyn, Poland. .,Foundation for the Nerve Cells Regeneration, Warszawska Str. 30, Mazury, 10-082, Olsztyn, Poland.
| | - Liliana Rytel
- Department of Internal Medicine and Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str 15, 10-718, Olsztyn, Poland
| | - Krystyna Makowska
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str. 13, 10-718, Olsztyn, Poland
| | - Sławomir Gonkowski
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str. 13, 10-718, Olsztyn, Poland
| |
Collapse
|
14
|
Lo SH, Niu HS, Cheng YZ, Niu CS, Cheng JT, Ku PM. Loperamide-induced Cardiac Depression Is Enhanced by Hyperglycemia: Evidence Relevant to Loperamide Abuse. Arch Med Res 2017; 48:64-72. [PMID: 28577871 DOI: 10.1016/j.arcmed.2017.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 01/16/2017] [Indexed: 12/18/2022]
|
15
|
Reiss D, Ceredig RA, Secher T, Boué J, Barreau F, Dietrich G, Gavériaux-Ruff C. Mu and delta opioid receptor knockout mice show increased colonic sensitivity. Eur J Pain 2016; 21:623-634. [PMID: 27748566 DOI: 10.1002/ejp.965] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Opiates act through opioid receptors to diminish pain. Here, we investigated whether mu (MOR) and delta (DOR) receptor endogenous activity assessed in the whole mouse body or in particular at peripheral receptors on primary nociceptive neurons, control colonic pain. METHODS We compared global MOR and DOR receptor knockout (KO) mice, mice with a conditional deletion of MOR and DOR in Nav1.8-positive nociceptive primary afferent neurons (cKO), and control floxed mice of both genders for visceral sensitivity. Visceromotor responses to colorectal distension (CRD) and macroscopic colon scores were recorded on naïve mice and mice with acute colitis induced by 3% dextran sodium sulphate (DSS) for 5 days. Transcript expression for opioid genes and cytokines was measured by quantitative RT-PCR. RESULTS Naïve MOR and DOR global KO mice show increased visceral sensitivity that was not observed in cKO mice. MOR and preproenkephalin (Penk) were the most expressed opioid genes in colon. MOR KO mice had augmented kappa opioid receptor and Tumour-Necrosis-Factor-α and diminished Penk transcript levels while DOR, preprodynorphin and Interleukin-1β were unchanged. Global MOR KO females had a thicker colon than floxed females. No alteration was detected in DOR mutant animals. A 5-day DSS treatment led to comparable hypersensitivity in the different mouse lines. CONCLUSION Our results suggest that mu and delta opioid receptor global endogenous activity but not activity at the peripheral Nav1.8 neurons contribute to visceral sensitivity in naïve mice, and that endogenous MOR and DOR tones were insufficient to elicit analgesia after 5-day DSS-induced colitis. SIGNIFICANCE Knockout mice for mu and delta opioid receptor have augmented colon sensitivity in the CRD assay. It shows endogenous mu and delta opioid analgesia that may be explored as potential targets for alleviating chronic intestinal pain.
Collapse
Affiliation(s)
- D Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France
| | - R A Ceredig
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Institut des Neurosciences Cellulaires et Intégratives INCI, UPR3212, Strasbourg, France
| | - T Secher
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - J Boué
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - F Barreau
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - G Dietrich
- Institut de Recherche en Santé Digestive IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - C Gavériaux-Ruff
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Centre National de la Recherche Scientifique, UMR7104, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France.,Ecole Supérieure de Biotechnologie de Strasbourg, Université de Strasbourg, France
| |
Collapse
|
16
|
Broad J, Maurel D, Kung VWS, Hicks GA, Schemann M, Barnes MR, Kenakin TP, Granier S, Sanger GJ. Human native kappa opioid receptor functions not predicted by recombinant receptors: Implications for drug design. Sci Rep 2016; 6:30797. [PMID: 27492592 PMCID: PMC4974614 DOI: 10.1038/srep30797] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/11/2016] [Indexed: 12/14/2022] Open
Abstract
If activation of recombinant G protein-coupled receptors in host cells (by drugs or other ligands) has predictive value, similar data must be obtained with native receptors naturally expressed in tissues. Using mouse and human recombinant κ opioid receptors transfected into a host cell, two selectively-acting compounds (ICI204448, asimadoline) equi-effectively activated both receptors, assessed by measuring two different cell signalling pathways which were equally affected without evidence of bias. In mouse intestine, naturally expressing κ receptors within its nervous system, both compounds also equi-effectively activated the receptor, inhibiting nerve-mediated muscle contraction. However, whereas ICI204448 acted similarly in human intestine, where κ receptors are again expressed within its nervous system, asimadoline was inhibitory only at very high concentrations; instead, low concentrations of asimadoline reduced the activity of ICI204448. This demonstration of species-dependence in activation of native, not recombinant κ receptors may be explained by different mouse/human receptor structures affecting receptor expression and/or interactions with intracellular signalling pathways in native environments, to reveal differences in intrinsic efficacy between receptor agonists. These results have profound implications in drug design for κ and perhaps other receptors, in terms of recombinant-to-native receptor translation, species-dependency and possibly, a need to use human, therapeutically-relevant, not surrogate tissues.
Collapse
Affiliation(s)
- John Broad
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Damien Maurel
- Institut de Génomique Fonctionnelle, Dépt de Pharmacologie Moléculaire, UMR 5203 CNRS-U 661 INSERM, Univ Montpellier I &II, 141, 34094 Montpellier, France
| | - Victor W S Kung
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Gareth A Hicks
- Tioga Pharmaceuticals, 9393 Towne Centre Drive, Suite 200, San Diego, California, USA
| | - Michael Schemann
- Human Biology, TU München, D-85350 Freising-Weihenstephan, Germany
| | - Michael R Barnes
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Terrence P Kenakin
- Dept of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sébastien Granier
- Institut de Génomique Fonctionnelle, Dépt de Pharmacologie Moléculaire, UMR 5203 CNRS-U 661 INSERM, Univ Montpellier I &II, 141, 34094 Montpellier, France
| | - Gareth J Sanger
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| |
Collapse
|
17
|
Li G, Low PS. Synthesis and evaluation of a ligand targeting the μ and δ opioid receptors for drug delivery to lung cancer. Bioorg Med Chem Lett 2016; 27:2074-2078. [PMID: 28291693 DOI: 10.1016/j.bmcl.2016.06.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/24/2016] [Accepted: 06/24/2016] [Indexed: 01/21/2023]
Abstract
A well-established approach to developing new imaging agents and treatments for cancer begins with the recognition of receptors that are overexpressed in cancer cells. Ideally, these same receptors would also be absent, or minimally expressed, in healthy tissue. The mu (μ) and delta (δ) opioid receptors (MOR and DOR respectively) match these criteria, with expression in cancer cells that is higher than primary lung epithelial cells. Naltrexone is a drug approved by the U.S. Food and Drug Administration (FDA) for treatment of alcohol dependence or prevention of relapse from opioid addiction. Since naltrexone binds with high affinity to both MOR and DOR, it was selected as the platform for development of novel ligands capable of delivering a cytotoxic payload to non-small cell lung cancer (NSCLC). This study outlines the synthesis of two ligands, with peptide or PEG linkers that were synthesized from 6-amino-naltrexone and conjugated with rhodamine dye or 99mTc for in vitro imaging, binding affinity or in vivo imaging and biodistribution studies. Transfected HEK cells were used as a model system for over-expression of the μ-opioid receptor (MOR) or the δ-opioid receptor (DOR). Naltrexone and naltrindole were used as competition for MOR and DOR respectively during the binding affinity studies. Mice bearing a xenograft of HEK cells transfected with μ (HEK-mu) or δ (HEK-delta) opioid receptors were the animal model used for PET imaging and in vivo biodistribution studies. Although the binding affinity studies were encouraging, the biodistribution data for the selected conjugates lacked sufficient specificity. These conjugates were abandoned from further development but information about their synthesis may be valuable to other laboratories working in this field.
Collapse
Affiliation(s)
- Guo Li
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette 47907, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, 720 Clinic Drive, West Lafayette 47907, USA
| |
Collapse
|
18
|
Opioid κ Receptors as a Molecular Target for the Creation of a New Generation of Analgesic Drugs. Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1388-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
19
|
First isolation and antinociceptive activity of a lipid transfer protein from noni ( Morinda citrifolia ) seeds. Int J Biol Macromol 2016; 86:71-9. [DOI: 10.1016/j.ijbiomac.2016.01.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 01/07/2016] [Accepted: 01/08/2016] [Indexed: 01/16/2023]
|
20
|
Role of DOR-β-arrestin1-Bcl2 signal transduction pathway and intervention effects of oxymatrine in ulcerative colitis. ACTA ACUST UNITED AC 2014; 34:815-820. [DOI: 10.1007/s11596-014-1358-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/27/2013] [Indexed: 11/25/2022]
|
21
|
Novel orally available salvinorin A analog PR-38 protects against experimental colitis and reduces abdominal pain in mice by interaction with opioid and cannabinoid receptors. Biochem Pharmacol 2014; 92:618-26. [PMID: 25265540 DOI: 10.1016/j.bcp.2014.09.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/17/2014] [Accepted: 09/18/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Salvinorin A (SA) is a potent anti-inflammatory diterpene isolated from the Mexican plant S. divinorum. Recently we showed that the novel SA analog, PR-38 has an inhibitory effect on mouse gastrointestinal (GI) motility mediated by opioid and cannabinoid (CB) receptors. The aim of the study was to characterize possible anti-inflammatory and antinociceptive action of PR-38 in the mouse GI tract. METHODS Macro- and microscopic colonic damage scores and myeloperoxidase activity were determined after intraperitoneal (i.p.), intracolonic (i.c.), and per os (p.o.) administration of PR-38 in the trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) models of colitis in mice. Additionally, MOP, KOP and CB1 protein expression was determined using Western blot analysis of mouse colon samples. The antinociceptive effect of PR-38 was examined based on the number of behavioral responses to i.c. instillation of mustard oil (MO). RESULTS The i.p. (10 mg/kg, twice daily), i.c. (10 mg/kg, twice daily) and p.o. (20 mg/kg, once daily) administration of PR-38 significantly attenuated TNBS- and DSS-induced colitis in mice. The effect of PR-38 was partially blocked by the KOP antagonist nor-binaltorphimine and CB1 antagonist AM 251. Western blot analysis showed a significant increase of MOP, KOP and CB1 receptor expression during colonic inflammation, which was reversed to the control levels by the administration of PR-38. PR-38 significantly decreased the number of pain responses after i.c. instillation of MO in the TNBS-treated mice. CONCLUSIONS Our results suggest that PR-38 has the potential to become a valuable anti-inflammatory and analgesic therapeutic for the treatment of GI inflammation.
Collapse
|
22
|
Fan H, Liu XX, Zhang LJ, Hu H, Tang Q, Duan XY, Zhong M, Shou ZX. Intervention effects of QRZSLXF, a Chinese medicinal herb recipe, on the DOR-β-arrestin1-Bcl2 signal transduction pathway in a rat model of ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:88-97. [PMID: 24637189 DOI: 10.1016/j.jep.2014.03.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 03/06/2014] [Accepted: 03/09/2014] [Indexed: 05/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qingre Zaoshi Liangxue Fang (QRZSLXF) is a Chinese medicinal herb recipe that is commonly prescribed for the treatment of ulcerative colitis. It includes 5 quality assured herbs: Sophora flavescens Aiton., Baphicacanthus cusia (Nees) Bremek., Bletilla striata Rchb.f., Glycyrrhiza uralensis Fisch. and Coptis chinensis Franch. The main phytochemical ingredient of QRZSLXF includes ammothamnine, sophocarpidine, liquiritin, berberine and indirubin. QRZSLXF has been clinically proven for use in the treatment of ulcerative colitis for over twenty years. In the past ten years, research has confirmed the therapeutic effect of QRZSLXF in ulcerative colitis and partially revealed its mechanism of action. Here, we further reveal the therapeutic mechanism of QRZSLXF in ulcerative colitis. To investigate the role of the DOR-β-arrestin1-Bcl-2 signal transduction pathway in ulcerative colitis and to determine the effects of QRZSLXF on this signal transduction pathway. MATERIALS AND METHODS Eighty-four Sprague-Dawley rats were randomly divided into six groups: normal control group, model group, mesalazine group, and QRZSLXF high-dose, medium-dose group and low-dose groups (n=14). Experimental colitis was induced by trinitrobenzenesulfonic acid (TNBS) in each group, except the normal control group. After modeling, bloody stool, mental state and diarrhea were observed and recorded. Two rats were randomly selected from the model groups adfnd sacrificed on day 3 to observe pathological changes in the colon tissue by microscopy. The rats in the QRZSLXF-treated groups received intramuscular injections of different concentrations of QRZSLXF for 15 days. The rats in the mesalazine group were treated with mesalazine solution (0.5 g/kg/day) by gastric lavage for 15 days. The rats in the normal control group and the model group were treated with 3 mL water by gastric lavage for 15 days. On the 16th day, after fasting for 24 h, the remaining rats were sacrificed and their colon tissues were used to detect the mRNA and protein expressions of DOR, β-arrestin1 and Bcl-2 by Real-time PCR and immunohistochemistry, respectively. Histological changes in the colon tissues were also examined. RESULTS AND CONCLUSIONS The expressions of DOR, β-arrestin1 and Bcl-2 were significantly different among the four groups. The expressions of DOR, β-arrestin1 and Bcl-2 protein and mRNA were significantly increased in the model group compared with the other groups (P<0.05). In contrast to the model group, the expressions of DOR, β-arrestin1 and Bcl-2 were significantly decreased in the mesalazine group and the groups that received different doses of QRZSLXF (P<0.05), and there were no statistically significant differences among the mesalazine and QRZSLXF-treated groups (P>0.05). This study indicates that the DOR-beta-arrestin1-Bcl-2 signal transduction pathway may participate in the pathologic course of ulcerative colitis. Moreover, QRZSLXF could attenuate ulcerative colitis by regulating the DOR-β-arrestin1-Bcl-2 signal transduction pathway.
Collapse
Affiliation(s)
- Heng Fan
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Xing-xing Liu
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Li-juan Zhang
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Hui Hu
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Qing Tang
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Xue-yun Duan
- Xueyun Duan, Department of Pharmacy, the Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan 430061, Hubei Province, China
| | - Min Zhong
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | - Zhe-xing Shou
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| |
Collapse
|
23
|
Loriga G, Lazzari P, Ruiu S, Marchese G, Manca I, Casu GL, Dessì C, Pinna GA, Asproni B, Murineddu G. Synthesis and biological evaluation of novel delta (δ) opioid receptor ligands with diazatricyclodecane skeletons. Eur J Med Chem 2013; 69:413-26. [PMID: 24090913 DOI: 10.1016/j.ejmech.2013.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 12/01/2022]
Abstract
Considering the interesting pharmacological profile of the delta (δ) selective opioid agonist compound SNC-80, conformationally constrained analogs containing two diazatricyclodecane ring systems in place of dimethylpiperazine core motif were synthesized. The compounds showed subnanomolar or low nanomolar δ opioid receptor binding affinity. Depending upon the substituents on the diazatricyclodecane ring, these compounds displayed varying selectivity for δ opioid receptor over μ and κ receptors. Amongst the novel compounds, 1Aa showed the more interesting biological profile, with higher δ affinity and selectivity compared to SNC-80. The δ receptor agonist profile and antinociceptive activity of 1Aa were confirmed using ex-vivo (isolated mouse vas deferens) and in vivo (tail flick) assays.
Collapse
Affiliation(s)
- Giovanni Loriga
- C.N.R. Istituto di Farmacologia Traslazionale, UOS Cagliari, Edificio 5, Loc. Piscinamanna, 09010 Pula, CA, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chen HJ, Xie WY, Hu F, Zhang Y, Wang J, Wang Y. Disruption of δ-opioid receptor phosphorylation at threonine 161 attenuates morphine tolerance in rats with CFA-induced inflammatory hypersensitivity. Neurosci Bull 2012; 28:182-92. [PMID: 22466129 DOI: 10.1007/s12264-012-1216-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Our previous study identified Threonine 161 (Thr-161), located in the second intracellular loop of the δ-opioid receptor (DOR), as the only consensus phosphorylation site for cyclin-dependent kinase 5 (Cdk5). The aim of this study was to assess the function of DOR phosphorylation by Cdk5 in complete Freund's adjuvant (CFA)-induced inflammatory pain and morphine tolerance. METHODS Dorsal root ganglion (DRG) neurons of rats with CFA-induced inflammatory pain were acutely dissociated and the biotinylation method was used to explore the membrane localization of phosphorylated DOR at Thr-161 (pThr-161-DOR), and paw withdrawal latency was measured after intrathecal delivery of drugs or Tat-peptide, using a radiant heat stimulator in rats with CFA-induced inflammatory pain. RESULTS Both the total amount and the surface localization of pThr-161-DOR were significantly enhanced in the ipsilateral DRG following CFA injection. Intrathecal delivery of the engineered Tat fusion-interefering peptide corresponding to the second intracellular loop of DOR (Tat-DOR-2L) increased inflammatory hypersensitivity, and inhibited DOR- but not µ-opioid receptor-mediated spinal analgesia in CFA-treated rats. However, intrathecal delivery of Tat-DOR-2L postponed morphine antinociceptive tolerance in rats with CFA-induced inflammatory pain. CONCLUSION Phosphorylation of DOR at Thr-161 by Cdk5 attenuates hypersensitivity and potentiates morphine tolerance in rats with CFA-induced inflammatory pain, while disruption of the phosphorylation of DOR at Thr-161 attenuates morphine tolerance.
Collapse
Affiliation(s)
- Hai-Jing Chen
- Neuroscience Research Institute, Peking University, Department of Neurobiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | | | | | | | | | | |
Collapse
|
25
|
Fichna J, Dicay M, Lewellyn K, Janecka A, Zjawiony JK, MacNaughton WK, Storr MA. Salvinorin A has antiinflammatory and antinociceptive effects in experimental models of colitis in mice mediated by KOR and CB1 receptors. Inflamm Bowel Dis 2012; 18:1137-45. [PMID: 21953882 DOI: 10.1002/ibd.21873] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/04/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND Salvinorin A (SA) has a potent inhibitory action on mouse gastrointestinal (GI) motility and ion transport, mediated primarily by kappa-opioid receptors (KOR). The aim of the present study was to characterize possible antiinflammatory and antinociceptive effects of SA in the GI tract of mice. METHODS Colonic damage scores and myeloperoxidase activity were determined after intraperitoneal (i.p.), intracolonic (i.c.), and oral (p.o.) administration of SA using the trinitrobenzene sulfonic acid (TNBS) and dextran sodium sulfate (DSS) models of colitis in mice. Additionally, KOR, cannabinoid (CB)1, and CB2 western blot analysis of colon samples was performed. The antinociceptive effect of SA was examined based on the number of behavioral responses to i.c. instillation of mustard oil (MO). RESULTS The i.p. (3 mg/kg, twice daily) and p.o. (10 mg/kg, twice daily) administration of SA significantly attenuated TNBS and DSS colitis in mice. The effect of SA was blocked by KOR antagonist nor-binaltorphimine (10 mg/kg, i.p.). Western blot analysis showed no influence of SA on KOR, CB1, or CB2 levels. SA (3 mg/kg, i.p. and 10 mg/kg, i.c.) significantly decreased the number of pain responses after i.c. instillation of MO in the vehicle- and TNBS-treated mice. The antinociceptive action of SA was blocked by KOR and CB1 antagonists. The analgesic effect of i.c. SA was more potent in TNBS-treated mice compared to controls. CONCLUSIONS Our results suggest that the drugs based on the structure of SA have the potential to become valuable antiinflammatory or analgesic therapeutics for the treatment of GI diseases.
Collapse
Affiliation(s)
- Jakub Fichna
- Snyder Institute of Infection, Immunity and Inflammation (III), University of Calgary, AB, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Gabrilovac J, Čupić B, Zapletal E, Brozovic A. IFN-γ up-regulates kappa opioid receptors (KOR) on murine macrophage cell line J774. J Neuroimmunol 2012; 245:56-65. [DOI: 10.1016/j.jneuroim.2012.02.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Revised: 02/06/2012] [Accepted: 02/08/2012] [Indexed: 11/29/2022]
|
27
|
Mangel AW, Hicks GA. Asimadoline and its potential for the treatment of diarrhea-predominant irritable bowel syndrome: a review. Clin Exp Gastroenterol 2012; 5:1-10. [PMID: 22346361 PMCID: PMC3278196 DOI: 10.2147/ceg.s23274] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a multifactorial condition with principal symptoms of pain and altered bowel function. The kappa-opioid agonist asimadoline is being evaluated in Phase III as a potential treatment for IBS. Asimadoline, to date, has shown a good safety profile and the target Phase III population - diarrhea-predominant IBS patients with at least moderate pain - was iteratively determined in a prospective manner from a Phase II dose-ranging study. The clinical data in support of this population are reviewed in this article. Furthermore, the scientific rationale for the use of asimadoline in the treatment of IBS is reviewed. Considering the high patient and societal burdens of IBS, new treatments for IBS represent therapeutic advances.
Collapse
|
28
|
Delta opioid receptor analgesia: recent contributions from pharmacology and molecular approaches. Behav Pharmacol 2011; 22:405-14. [PMID: 21836459 DOI: 10.1097/fbp.0b013e32834a1f2c] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Delta opioid receptors represent a promising target for the development of novel analgesics. A number of tools have been developed recently that have significantly improved our knowledge of δ receptor function in pain control. These include several novel δ agonists with potent analgesic properties, and genetic mouse models with targeted mutations in the δ opioid receptor gene. Also, recent findings have further documented the regulation of δ receptor function at cellular level, which impacts on the pain-reducing activity of the receptor. These regulatory mechanisms occur at transcriptional and post-translational levels, along agonist-induced receptor activation, signaling and trafficking, or in interaction with other receptors and neuromodulatory systems. All these tools for in-vivo research, and proposed mechanisms at molecular level, have tremendously increased our understanding of δ receptor physiology, and contribute to designing innovative strategies for the treatment of chronic pain and other diseases such as mood disorders.
Collapse
|
29
|
de Freitas Pires A, Assreuy AMS, Lopes ÉAB, Celedônio NR, Soares CEA, Rodrigues NVFC, Sousa PL, Benevides RG, Nagano CS, Cavada BS, Leal-Cardoso JH, Coelho-de-Souza AN, Santos CF. Opioid-like antinociceptive effects of oral administration of a lectin purified from the seeds of Canavalia brasiliensis. Fundam Clin Pharmacol 2011; 27:201-9. [PMID: 21895762 DOI: 10.1111/j.1472-8206.2011.00987.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The objective of this study was to evaluate the antinociceptive effects of a lectin from Canavalia brasiliensis (ConBr) when administered orally to murine models of chemical and thermal nociception. ConBr up to 100 mg/kg produced significant and dose-dependent antinociceptive effects: 81% reduction in abdominal writhing induced by 0.6% acetic acid; 26 and 52% reduction in early- and late-stage paw licking, respectively, induced by 2.5% formalin; and 155% increase in reaction latency (heightened thermal pain threshold). In all models, the antinociceptive effect was reversed by the lectin-binding carbohydrate α-d-methyl-mannoside and by the nonselective opioid antagonist naloxone. The antinociceptive effect observed in the formalin test was inhibited by the δ-selective antagonist naltrindole and the κ-selective antagonist nor-binaltorphimine but not by the μ-selective antagonist cyprodime. In conclusion, when administered orally to Swiss mice, the ConBr lectin displayed antinociceptive activity, both peripheral and central, mediated by the opioid system and involving δ-and κ-receptors and the lectin domain.
Collapse
Affiliation(s)
- Alana de Freitas Pires
- Instituto Superior de Ciências Biomédicas, Universidade Estadual do Ceará, Av. Paranjana 1700, 60.740-000, Fortaleza, Ceará, Brazil
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Olesen AE, Staahl C, Arendt-Nielsen L, Drewes AM. Different effects of morphine and oxycodone in experimentally evoked hyperalgesia: a human translational study. Br J Clin Pharmacol 2011; 70:189-200. [PMID: 20653672 DOI: 10.1111/j.1365-2125.2010.03700.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT * Previous studies using short-lasting experimental pain stimulations in healthy volunteers have shown differences in opioid effects regarding visceral pain stimulations. However, these differences can be more pronounced in patients due to a sensitized pain system. Therefore, the aim of the present study was to mimic the clinical situation by investigating opioid effects on experimental pain in healthy volunteers after experimentally evoked hyperalgesia. WHAT THIS STUDY ADDS? * We now know that morphine and oxycodone exerts different effects in the sensitized pain system as we found a greater analgesic effect of oxycodone in response to skin, muscle and oesophageal pain stimulation. This supports clinicians' experiences that oxycodone can be superior to morphine in the treatment of some pain conditions. The evoked hyperalgesia bridged findings from studies in healthy volunteers to patients, and new fundamental knowledge on different analgesic effects in hyperalgesia was found. AIM Similar analgesics may have different analgesic potencies especially in patients in whom the pain system is sensitized. The aim was to investigate different opioid effects on experimental pain after the sensitized pain system was mimicked evoking hyperalgesia in healthy volunteers. METHODS Twenty-four healthy volunteers were randomized to treatment with morphine (30 mg orally) and oxycodone (15 mg orally) or placebo in a double-blind crossover study. Hyperalgesia was induced by oesophageal perfusion with acid and capsaicin. Several exploratory endpoints were studied using skin heat, muscle pressure and oesophageal mechanical, heat and electrical stimulation. Effects on pain from deeper structures were considered most important. RESULTS Different analgesic potencies were found. Oxycodone had a greater analgesic effect than morphine attenuating pain from: (i) heat stimulation of skin (P= 0.016); difference between the means of 0.39 degrees C, 95% CI 0.22, 2.09. (ii) muscle pressure (P < 0.001); difference between the means of 11.93kPa, 95% CI 5.4, 18.5. (iii) oesophageal heat stimulation (P < 0.001); difference between the means of 38.54 cm(2), 95% CI 15.37, 61.71 and (iv) oesophageal electrical stimulation (P= 0.016); difference between the means of 6.69mA, 95% CI 1.23, 12.13. CONCLUSION After sensitization of the pain system different analgesic potencies of morphine and oxycodone were found in response to skin, muscle and oesophageal pain stimulation, in which oxycodone had a greater effect. As similar differential analgesic potencies of the two opioids have been found in patients with chronic pain, the experimental hyperalgesia model bridged findings from studies in healthy volunteers to patients.
Collapse
Affiliation(s)
- Anne Estrup Olesen
- Mech-Sense, Department of Gastroenterology, Aalborg Hospital, Aarhus University Hospital, Aalborg, Denmark.
| | | | | | | |
Collapse
|
31
|
Olesen AE, Staahl C, Brock C, Arendt-Nielsen L, Drewes AM. Evoked Human Oesophageal Hyperalgesia: A Potential Tool for Analgesic Evaluation? Basic Clin Pharmacol Toxicol 2009; 105:126-36. [DOI: 10.1111/j.1742-7843.2009.00422.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
32
|
Kokrashvili Z, Rodriguez D, Yevshayeva V, Zhou H, Margolskee RF, Mosinger B. Release of endogenous opioids from duodenal enteroendocrine cells requires Trpm5. Gastroenterology 2009; 137:598-606, 606.e1-2. [PMID: 19272386 PMCID: PMC2717179 DOI: 10.1053/j.gastro.2009.02.070] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 02/14/2009] [Accepted: 02/23/2009] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Enteroendocrine cells, the largest and most diverse population of mammalian endocrine cells, comprise a number of different cell types in the gut mucosa that produce, store, and secrete small molecules, peptides, and/or larger proteins that regulate many aspects of gut physiology. Little is known about less typical endocrine cells in the intestinal mucosa that do not contain secretory granules, such as brush or caveolated cells. We studied a subset of these enteroendocrine cells in duodenum that produce several peptides, including endogenous opioids, and that also express the Trpm5 cation channel. METHODS We studied expression patterns of Trpm5 and other molecules by immunohistochemical and enzyme-linked immunosorbent assay analyses of intestinal tissues from transgenic mice that express green fluorescent protein from the Trpm5 promoter, as well as wild-type and Trpm5-null mice. RESULTS We describe a type of enteroendocrine cell in mouse duodenum that is defined by the presence of Trpm5 and that does not contain typical secretory granules yet expresses endogenous opioids (beta-endorphin and Met-enkephalin) and uroguanylin in apical compartments close to the lumen of the gut. CONCLUSIONS Solitary chemosensory cells that coexpress beta-endorphin, Met-enkephalin, uroguanylin, and Trpm5 exist in mouse duodenum. These cells are likely to secrete the bioactive peptides into the intestinal lumen in response to dietary factors; release of the opioid peptides requires the Trpm5 ion channel.
Collapse
Affiliation(s)
| | | | | | | | | | - Bedrich Mosinger
- Department of Neuroscience, 1425 Madison Ave, New York, NY 10029. E-mail: , Telephone: 212 659 8695, Fax: 212849 2599
| |
Collapse
|
33
|
Vats ID, Snehlata, Nath M, Pasha MAQ, Pasha S. Effect of chronic intra-peritoneally administered chimeric peptide of met-enkephalin and FMRFa-[D-Ala2]YFa-on antinociception and opioid receptor regulation. Eur J Pain 2009; 14:295.e1-9. [PMID: 19560378 DOI: 10.1016/j.ejpain.2009.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 05/16/2009] [Accepted: 05/23/2009] [Indexed: 11/29/2022]
Abstract
The physiological role of NPFF/FMRFa family of peptides is complex and exact mechanism of action of these peptides is not yet completely understood. In same line of scrutiny, previously we reported an enzymatically stable chimeric analog of YGGFMKKKFMRFamide (YFa) i.e., [D-Ala(2)]YAGFMKKKFMRFamide ([D-Ala(2)]YFa) which have a role in antinociception and modulatory effect on opioid analgesia. In continuation, presently we investigated using tail-flick test whether [D-Ala(2)]YFa on systemic administration induced any antinociception in rats and if so then which specific opioid receptor(s) mu, delta or kappa mediated it. Further, the antinociceptive effect of [D-Ala(2)]YFa on 6 days chronic intra-peritoneal (i.p.) treatment in rats was examined and finally, effect of this chronic treatment on the differential expression of opioid receptors was assessed. [D-Ala(2)]YFa on i.p. administration induced dose dependent antinociception which was mainly mediated by delta (DOR) and partially by mu (MOR) and kappa (KOR) opioid receptors. Moreover, its antinociceptive effect remained comparable throughout the chronic treatment even during insufficient availability of DOR1. Importantly, during this treatment the mRNA expression of all three opioid receptors (MOR1, KOR1 and DOR1) was increased as assessed by real-time RTPCR though subsequent western blot analysis revealed a selective increase in the protein level of DOR1, only. Thus, pharmacological behavior of [d-Ala(2)]YFa suggests that competency of an opioid agonist to bind with multiple opioid receptors may enhance its potency to induce tolerance free analgesia.
Collapse
Affiliation(s)
- Ishwar Dutt Vats
- Peptide Synthesis Laboratory, Institute of Genomics and Integrative Biology, Delhi, India
| | | | | | | | | |
Collapse
|
34
|
Josan JS, Morse DL, Xu L, Trissal M, Baggett B, Davis P, Vagner J, Gillies RJ, Hruby VJ. Solid-phase synthetic strategy and bioevaluation of a labeled delta-opioid receptor ligand Dmt-Tic-Lys for in vivo imaging. Org Lett 2009; 11:2479-82. [PMID: 19445485 PMCID: PMC2756606 DOI: 10.1021/ol900200k] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A general solid-phase synthetic strategy is developed to prepare fluorescent and/or lanthanide-labeled derivatives of the delta-opioid receptor (deltaOR) ligand H-Dmt-Tic-Lys(R)-OH. The high delta-OR affinity (K(i) = 3 nM) and desirable in vivo characteristics of the Cy5 derivative 1 suggest its usefulness for structure-function studies and receptor localization and as a high-contrast noninvasive molecular marker for live imaging ex vivo or in vivo.
Collapse
Affiliation(s)
- Jatinder S Josan
- Department of Chemistry, BIO5 Institute, The University of Arizona, Tucson, Arizona 85721, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Balboni G, Fiorini S, Baldisserotto A, Trapella C, Sasaki Y, Ambo A, Marczak ED, Lazarus LH, Salvadori S. Further studies on lead compounds containing the opioid pharmacophore Dmt-Tic. J Med Chem 2008; 51:5109-17. [PMID: 18680274 PMCID: PMC2812024 DOI: 10.1021/jm800587e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Some reference opioids containing the Dmt-Tic pharmacophore, especially the delta agonists H-Dmt-Tic-Gly-NH-Ph (1) and H-Dmt-Tic-NH-(S)CH(CH2-COOH)-Bid (4) (UFP-512) were evaluated for the influence of the substitution of Gly with aspartic acid, its chirality, and the importance of the -NH-Ph and N(1)H-Bid hydrogens in the inductions of delta agonism. The results provide the following conclusions: (i) Asp increases delta selectivity by lowering the mu affinity; (ii) -NH-Ph and N(1)H-Bid nitrogens methylation transforms the delta agonists into delta antagonists; (iii) the substitution of Gly with L-Asp/D-Asp in the delta agonist H-Dmt-Tic-Gly-NH-Ph gave delta antagonists; the same substitution in the delta agonist H-Dmt-Tic-NH-CH2-Bid yielded more selective agonists, H-Dmt-Tic-NH-(S)CH(CH2-COOH)-Bid and H-Dmt-Tic-NH-(R)CH(CH2-COOH)-Bid; (iv) L-Asp seems important only in functional bioactivity, not in receptor affinity; (v) H-Dmt-Tic-NH-(S)CH(CH2-COOH)-Bid(N(1)-Me) (10) evidenced analgesia similar to 4, which was reversed by naltrindole only in the tail flick. 4 and 10 had opposite behaviours in mice; 4 caused agitation, 10 gave sedation and convulsions.
Collapse
Affiliation(s)
- Gianfranco Balboni
- Department of Toxicology, University of Cagliari, I-09124, Cagliari, Italy
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, I-44100 Ferrara, Italy
| | - Stella Fiorini
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, I-44100 Ferrara, Italy
| | - Anna Baldisserotto
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, I-44100 Ferrara, Italy
| | - Claudio Trapella
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, I-44100 Ferrara, Italy
| | - Yusuke Sasaki
- Department of Pharmacology, Tohoku Pharmaceutical University, 4-1, Komatsushima 4-chome, Aoba-Ku, Sendai 981-8558, Japan
| | - Akihiro Ambo
- Department of Pharmacology, Tohoku Pharmaceutical University, 4-1, Komatsushima 4-chome, Aoba-Ku, Sendai 981-8558, Japan
| | - Ewa D. Marczak
- Medicinal Chemistry Group, Laboratory of Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Lawrence H. Lazarus
- Medicinal Chemistry Group, Laboratory of Pharmacology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Severo Salvadori
- Department of Pharmaceutical Sciences and Biotechnology Center, University of Ferrara, I-44100 Ferrara, Italy
| |
Collapse
|
36
|
Schmidt J, Stoffels B, Nazir A, Dehaven-Hudkins DL, Bauer AJ. Alvimopan and COX-2 inhibition reverse opioid and inflammatory components of postoperative ileus. Neurogastroenterol Motil 2008; 20:689-99. [PMID: 18266613 DOI: 10.1111/j.1365-2982.2007.01078.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our objective was to investigate the therapeutic potential of peripheral opioid antagonism with alvimopan and anti-inflammatory cyclooxygenase 2 (COX-2) inhibition in an animal model of postoperative ileus with pain management. Intestinal manipulation was conducted in mice and rats with or without postoperative morphine injection. Rodents were orally fed non-digestible fluorescein (FITC)-labelled dextran and transit measured after a period of 90 min. The immunomodulatory effects of morphine and alvimopan were determined on nitric oxide released from the organ cultured muscularis externa. Surgical manipulation of the intestine resulted in a delay in gastrointestinal transit after 24 h that worsened with exogenous morphine. Alvimopan did not significantly alter transit of control or manipulated animals, but significantly antagonized the transit delaying effects of morphine. However, when the inflammatory component was robust enough to obscure a further opioid induced delay in gastrointestinal transit, alvimopan ceased to be effective in improving postoperative intestinal function. Cyclooxygenase 2 inhibition significantly diminished the inflammatory component of postoperative ileus. Surgical manipulation resulted in an increased release of nitric oxide from the inflamed isolated muscularis externa in 24-h organ culture which was not altered by morphine or alvimopan. Two distinct mechanisms exist which participate in postoperative bowel dysfunction: a local inflammatory response which is antagonized by COX-2 inhibition, and a morphine-induced alteration in neural function which can be blocked with alvimopan.
Collapse
Affiliation(s)
- J Schmidt
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
37
|
Cannabidiol, extracted from Cannabis sativa, selectively inhibits inflammatory hypermotility in mice. Br J Pharmacol 2008; 154:1001-8. [PMID: 18469842 DOI: 10.1038/bjp.2008.177] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Cannabidiol is a Cannabis-derived non-psychotropic compound that exerts a plethora of pharmacological actions, including anti-inflammatory, neuroprotective and antitumour effects, with potential therapeutic interest. However, the actions of cannabidiol in the digestive tract are largely unexplored. In the present study, we investigated the effect of cannabidiol on intestinal motility in normal (control) mice and in mice with intestinal inflammation. EXPERIMENTAL APPROACH Motility in vivo was measured by evaluating the distribution of an orally administered fluorescent marker along the small intestine; intestinal inflammation was induced by the irritant croton oil; contractility in vitro was evaluated by stimulating the isolated ileum, in an organ bath, with ACh. KEY RESULTS In vivo, cannabidiol did not affect motility in control mice, but normalized croton oil-induced hypermotility. The inhibitory effect of cannabidiol was counteracted by the cannabinoid CB1 receptor antagonist rimonabant, but not by the cannabinoid CB2 receptor antagonist SR144528 (N-[-1S-endo-1,3,3-trimethyl bicyclo [2.2.1] heptan-2-yl]-5-(4-chloro-3-methylphenyl)-1-(4-methylbenzyl)-pyrazole-3-carboxamide), by the opioid receptor antagonist naloxone or by the alpha2-adrenergic antagonist yohimbine. Cannabidiol did not reduce motility in animals treated with the fatty acid amide hydrolase (FAAH) inhibitor N-arachidonoyl-5-hydroxytryptamine, whereas loperamide was still effective. In vitro, cannabidiol inhibited ACh-induced contractions in the isolated ileum from both control and croton oil-treated mice. CONCLUSIONS AND IMPLICATIONS Cannabidiol selectively reduces croton oil-induced hypermotility in mice in vivo and this effect involves cannabinoid CB1 receptors and FAAH. In view of its low toxicity in humans, cannabidiol may represent a good candidate to normalize motility in patients with inflammatory bowel disease.
Collapse
|
38
|
Ryu EK, Wu Z, Chen K, Lazarus LH, Marczak ED, Sasaki Y, Ambo A, Salvadori S, Ren C, Zhao H, Balboni G, Chen X. Synthesis of a potent and selective (18)F-labeled delta-opioid receptor antagonist derived from the Dmt-Tic pharmacophore for positron emission tomography imaging. J Med Chem 2008; 51:1817-23. [PMID: 18311909 PMCID: PMC2667121 DOI: 10.1021/jm7014765] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Identification and pharmacological characterization of two new selective delta-opioid receptor antagonists, derived from the Dmt-Tic pharmacophore, of potential utility in positron emission tomography (PET) imaging are described. On the basis of its high delta selectivity, H-Dmt-Tic--Lys(Z)-OH (reference compound 1) is a useful starting point for the synthesis of (18)F-labeled compounds prepared by the coupling of N-succinimidyl 4-[ (18)F]fluorobenzoate ([(18)F]SFB) with Boc-Dmt-Tic--Lys(Z)-OH under slightly basic conditions at 37 degrees C for 15 min, deprotection with TFA, and HPLC purification. The total synthesis time was 120 min, and the decay-corrected radiochemical yield of [(18)F]- 1 was about 25-30% ( n = 5) starting from [(18)F]SFB ( n = 5) with an effective specific activity about 46 GBq/micromol. In vitro autoradiography studies showed prominent uptake of [ (18)F]- 1 in the striatum and cortex with significant blocking by 1 and UFP-501 (selective delta-opioid receptor antagonist), suggesting high specific binding of [(18)F]- 1 to delta-opioid receptors. Noninvasive microPET imaging studies revealed the absence of [(18)F]- 1 in rat brain, since it fails to cross the blood-brain barrier. This study demonstrates the suitability of [ (18)F]- 1 for imaging peripheral delta-opioid receptors.
Collapse
Affiliation(s)
- Eun Kyoung Ryu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5484, USA
| | - Zhanhong Wu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5484, USA
| | - Kai Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5484, USA
| | - Lawrence H. Lazarus
- Medicinal Chemistry Group, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ewa, D. Marczak
- Medicinal Chemistry Group, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Yusuke Sasaki
- Tohoku Pharmaceutical University, 4-1, Komatsushima 4-chome, Aoba-Ku, Sendai 981-8558, Japan
| | - Akihiro Ambo
- Tohoku Pharmaceutical University, 4-1, Komatsushima 4-chome, Aoba-Ku, Sendai 981-8558, Japan
| | - Severo Salvadori
- Department of Pharmaceutical Science and Biotechnology Center, University of Ferrara, I-44100, Ferrara, Italy
| | - Chuancheng Ren
- Department of Neurosurgery, Stanford University, Stanford, CA 94305-5327, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, CA 94305-5327, USA
| | - Gianfranco Balboni
- Department of Pharmaceutical Science and Biotechnology Center, University of Ferrara, I-44100, Ferrara, Italy
- Department of Toxicology, University of Cagliari, I-09124, Cagliari, Italy
| | - Xiaoyuan Chen
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, CA 94305-5484, USA
| |
Collapse
|
39
|
Sykes KT, White SR, Hurley RW, Mizoguchi H, Tseng LF, Hammond DL. Mechanisms responsible for the enhanced antinociceptive effects of micro-opioid receptor agonists in the rostral ventromedial medulla of male rats with persistent inflammatory pain. J Pharmacol Exp Ther 2007; 322:813-21. [PMID: 17494863 DOI: 10.1124/jpet.107.121954] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This study investigated three possible mechanisms by which the antinociceptive effects of the mu-opioid receptor (MOR) agonist [d-Ala(2),N-Me-Phe(4),Gly(5)-ol]-enkephalin (DAMGO) and the delta-opioid receptor (DOR) agonist [d-Ala(2),Glu(4)]-deltorphin (deltorphin II) (DELT), microinjected into the rostral ventromedial medulla (RVM), are enhanced in rats with persistent inflammatory injury. Radioligand binding determined that neither the B(max) nor the K(d) values of [(3)H]DAMGO differed in RVM membranes from rats that received an intraplantar injection of saline or complete Freund's adjuvant (CFA) in one hindpaw 4 h, 4 days, or 2 weeks earlier. Likewise, neither the EC(50) nor the E(max) value for DAMGO-induced stimulation of guanosine 5'-O-(3-[(35)S]thio)triphosphate ([(35)S]GTPgammaS) binding differed in the RVM of saline- or CFA-treated rats at any time point. Microinjection of fixed dose combinations of DAMGO and DELT in the RVM of naive rats indicated that these agonists interact synergistically to produce antinociception when DAMGO is present in equal or greater amounts than DELT and, additively, when DELT is the predominant component. Thus, unlike the periphery or spinal cord, potentiation of MOR-mediated antinociception does not entail an increase in MOR number, affinity, or coupling. Rather, the data are concordant with our proposal that potentiation results from a synergistic interaction of exogenous MOR agonist with DOR-preferring enkephalins whose levels are increased in CFA-treated rats (J Neurosci 21:2536-2545, 2001). Virtually no specific [(3)H]DELT binding nor stimulation of [(35)S]GTPgammaS binding by DELT was obtained in RVM membranes from CFA- or saline-treated rats at any time point. The mechanisms responsible for the potentiation of DELT-mediated antinociception remain to be elucidated.
Collapse
MESH Headings
- Analgesics/metabolism
- Analgesics/pharmacology
- Animals
- Cell Membrane/metabolism
- Dose-Response Relationship, Drug
- Drug Synergism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/metabolism
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Freund's Adjuvant/pharmacology
- Guanosine 5'-O-(3-Thiotriphosphate)/metabolism
- Hindlimb
- Hyperalgesia/etiology
- Hyperalgesia/metabolism
- Hyperalgesia/prevention & control
- Inflammation/chemically induced
- Inflammation/complications
- Inflammation/pathology
- Male
- Medulla Oblongata/chemistry
- Medulla Oblongata/drug effects
- Medulla Oblongata/metabolism
- Oligopeptides/pharmacology
- Pain/etiology
- Pain/metabolism
- Pain/prevention & control
- Pain Measurement/methods
- Rats
- Rats, Sprague-Dawley
- Reaction Time/drug effects
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/analysis
- Receptors, Opioid, mu/metabolism
Collapse
Affiliation(s)
- Kenneth T Sykes
- Department of Anesthesia, The University of Iowa, 200 Hawkins Dr. 6 JCP, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
40
|
Labuz D, Mousa SA, Schäfer M, Stein C, Machelska H. Relative contribution of peripheral versus central opioid receptors to antinociception. Brain Res 2007; 1160:30-8. [PMID: 17599812 DOI: 10.1016/j.brainres.2007.05.049] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 05/08/2007] [Accepted: 05/09/2007] [Indexed: 11/30/2022]
Abstract
Opioid effects are mediated by central and peripheral opioid receptors. Here we examine the relative contribution of each receptor population to antinociception elicited by systemically administered centrally penetrating opioids, and by loperamide (a peripherally restricted opioid). Nociception (abdominal writhes) was induced by intraperitoneally (i.p.) injected 0.6% acetic acid in mice. We analyzed opioid receptor expression in peritoneum by immunohistochemistry, antinociception after i.p. injected agonists at mu (morphine, loperamide)-, delta (SNC80)- and kappa (U50488)-receptors, and its reversibility by subcutaneously (s.c.) administered centrally penetrating antagonists beta-funaltrexamine (mu), naltrindole (delta) and nor-binaltorphimine (kappa), and by the peripherally restricted antagonist naloxone methiodide (NLXM). NLXM was also injected intracerebroventricularly (i.c.v.) before i.p. loperamide. Mu-, kappa- and, to a lesser degree, delta-receptors were expressed on peripheral nerve terminals in the peritoneum. The anatomical distribution of the opioid receptor staining was very similar to the staining for calcitonin gene-related peptide, a marker of sensory neurons. Morphine, U50488 and, to a lesser degree, SNC80 blocked acetic and acid induced writhes. These effects were reversed by beta-funaltrexamine, nor-binaltorphimine and naltrindole, respectively. NLXM (s.c.) reversed antinociceptive effects of morphine, SNC80 and U50488 by 57%, 80% and 47%, respectively. Loperamide (0.05 mg/kg)-induced antinociception was reversed by s.c. beta-funaltrexamine and NLXM. Loperamide (0.1 mg/kg)-induced antinociception was completely blocked by s.c. beta-funaltrexamine but was only attenuated (by 50%) by s.c. or i.c.v. NLXM. In conclusion, systemically administered centrally penetrating mu-, delta- and kappa-agonists produced a substantial part of antinociception through peripheral opioid receptors. Higher dose loperamide-induced antinociception involved also central opioid receptors.
Collapse
Affiliation(s)
- Dominika Labuz
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité-Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | | | | | | | | |
Collapse
|
41
|
Puehler W, Rittner HL, Mousa SA, Brack A, Krause H, Stein C, Schäfer M. Interleukin-1 beta contributes to the upregulation of kappa opioid receptor mrna in dorsal root ganglia in response to peripheral inflammation. Neuroscience 2006; 141:989-998. [PMID: 16782281 DOI: 10.1016/j.neuroscience.2006.03.078] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Revised: 03/22/2006] [Accepted: 03/30/2006] [Indexed: 01/13/2023]
Abstract
During local painful inflammation, axonal transport of opioid receptors from dorsal root ganglia toward the periphery is increased, associated with a higher receptor density and enhanced efficacy of opioid analgesics at the injured site. To examine whether this increase is related to transcription, mRNA of the kappa opioid receptor in lumbar dorsal root ganglia was quantified by real time light cycler polymerase chain reaction. In dorsal root ganglia of naive rats, kappa opioid receptor mRNA expression was three-fold higher than previously shown for delta opioid receptor and two times lower than mu opioid receptor mRNA, respectively. After induction of unilateral paw inflammation by Freund's complete adjuvant, kappa opioid receptor mRNA was significantly upregulated with a peak at 12 h in ipsilateral dorsal root ganglia. This effect could be mimicked by intraplantar injection of the proinflammatory cytokine interleukin-1 beta. Kappa opioid receptor mRNA upregulation lasted longer in interleukin-1 beta-treated rats compared with Freund's complete adjuvant-treated rats. Furthermore, a significant increase in kappa opioid receptor positive neurons was detected by immunohistochemistry 24 h after local injection of Freund's complete adjuvant or interleukin-1 beta. In Freund's complete adjuvant-induced inflammation, kappa opioid receptor upregulation was blocked by treatment with interleukin-1 receptor antagonist without changing the leukocyte infiltration in the paw. In conclusion, kappa opioid receptor mRNA and protein in dorsal root ganglia are upregulated in response to peripheral inflammation. This effect can be mimicked by a single local injection of interleukin-1 beta, and Freund's complete adjuvant-induced upregulation in kappa opioid receptor mRNA and protein can be prevented by treatment with interleukin-1 receptor antagonist. These data suggest that the peripheral production of the proinflammatory cytokine interleukin-1 beta is a specific inducer of kappa opioid receptor expression in the dorsal root ganglia.
Collapse
Affiliation(s)
- W Puehler
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany.
| | - H L Rittner
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | - S A Mousa
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | - A Brack
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | - H Krause
- Klinik für Urologie, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | - C Stein
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| | - M Schäfer
- Klinik für Anaesthesiologie und operative Intensivmedizin, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, D-12200 Berlin, Germany
| |
Collapse
|
42
|
Jiménez N, Puig MM, Pol O. Antiexudative effects of opioids and expression of kappa- and delta-opioid receptors during intestinal inflammation in mice: involvement of nitric oxide. J Pharmacol Exp Ther 2006; 316:261-70. [PMID: 16183704 DOI: 10.1124/jpet.105.091991] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The study evaluates the effects of kappa- (KOR), delta- (DOR), and mu-opioid receptor (MOR) agonists on the inhibition of plasma extravasation during acute and chronic intestinal inflammation in mice. The antiexudative effects of KOR and DOR agonists in animals treated with nitric oxide synthase (NOS) inhibitors and their protein levels in the gut (whole jejunum and mucosa) and spinal cord of mice with chronic intestinal inflammation were also measured. Inflammation was induced by the intragastric administration of one (acute) or two (chronic) doses of croton oil. Plasma extravasation was measured using Evans blue and protein levels by Western blot and immunoprecipitation. Plasma extravasation was significantly increased 2.7 times during chronic inflammation. The potency of the KOR agonist trans-3,4-dichloro-N-methyl-N-[2-(1-pyrrolydinyl)cyclohexyl]-benzeneazetamine (U50,488H) inhibiting plasma extravasation was enhanced 26.3 times during chronic compared with acute inflammation. [d-Pen(2),d-Pen(5)]-Enkephalin (DPDPE) (a DOR agonist) was also 11.8 times more potent during chronic inflammation, whereas the antiexudative effects of fentanyl (a MOR agonist) were not significantly altered. Receptor-specific antagonists reversed the effects. Protein levels of KOR and DOR in the whole jejunum and mucosa were significantly increased after chronic inflammation. Treatment with NOS inhibitors N(omega)-nitro-l-arginine methyl ester or l-N(6)-(1-iminoethyl)-lysine hydrochloride diminished plasma extravasation and inhibited the increased antiexudative effects of U50,488H and DPDPE during chronic intestinal inflammation. The data show that the enhanced antiexudative effects of KOR and DOR agonists could be related to an increased expression of KOR and DOR in the gut and that the release of nitric oxide may play a role augmenting the effects of opioids during chronic inflammation.
Collapse
Affiliation(s)
- Natalia Jiménez
- Laboratori de Neurofarmacologia Molecular, Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Edifici C-Z, 08193 Barcelona, Spain
| | | | | |
Collapse
|
43
|
Chadzinska M, Starowicz K, Scislowska-Czarnecka A, Bilecki W, Pierzchala-Koziec K, Przewlocki R, Przewlocka B, Plytycz B. Morphine-induced changes in the activity of proopiomelanocortin and prodynorphin systems in zymosan-induced peritonitis in mice. Immunol Lett 2005; 101:185-92. [PMID: 15979727 DOI: 10.1016/j.imlet.2005.05.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 05/16/2005] [Accepted: 05/26/2005] [Indexed: 11/16/2022]
Abstract
We have shown that supplementation of proinflammatory agent with a high dose of morphine not only abolishes inflammation-related pain symptoms but also inhibits influx of leukocytes to the inflamed peritoneal cavity. Present investigations focused on effects of morphine on proopiomelanocortin and prodynorphin systems during zymosan-induced peritonitis. Males of SWISS mice were ip injected with zymosan (Z, 40 mg/kg) or zymosan with morphine (ZM, 20 mg/kg). At time 0 (controls) and 4 and 24h after stimulation, peritoneal leukocytes (PTLs) were counted, PTL levels of opioid peptides (beta-endorphin and dynorphin) measured by radioimmunoassays, while mRNAs coding their respective precursors (POMC and PDYN) and receptors (MOR and KOR) determined by QRT-PCR. Influx of inflammatory PTLs, mainly PMNs, was significantly delayed by morphine co-injection. Total levels of beta-endorphin and dynorphin corresponded with PTL numbers, while levels per cell were similar in all groups except of beta-endorphin, decreased in ZM at 4h. Levels of both peptides in peritoneal fluid were increased in Z and ZM groups at 4h, while at 24h only in case of beta-endorphin in Z group. POMC was increased only in ZM group at 4h of peritonitis, while PDYN in both Z and ZM groups at the same time. MOR mRNA was increased 24h after injection in Z and ZM groups, while KOR mRNA was similar in all groups except of decrease in Z at 24h. In conclusion, endogenous opioids and their receptors are involved in zymosan-induced peritonitis and affected in various ways by morphine co-injection.
Collapse
Affiliation(s)
- M Chadzinska
- Department of Evolutionary Immunobiology, Institute of Zoology, Jagiellonian University, Cracow, Poland
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Börner C, Wöltje M, Höllt V, Kraus J. STAT6 transcription factor binding sites with mismatches within the canonical 5'-TTC...GAA-3' motif involved in regulation of delta- and mu-opioid receptors. J Neurochem 2005; 91:1493-500. [PMID: 15584925 DOI: 10.1111/j.1471-4159.2004.02846.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Opioid receptors are expressed in neuronal and immune cells and regulated in response to immunological processes. Herein, we demonstrate up-regulation of the delta-opioid receptor gene by interleukin-4 in immune cells (primary T and polymorphonuclear leukocytes, Jurkat E6 T cells), and in NG 108-15 neuronal cells. We identified an interleukin-4-responsive element at nt -671 on the murine gene promoter, to which the transcription factor STAT6 binds, as shown by reporter gene analysis and STAT6/DNA interaction studies in living cells with transcription factor decoy oligonucleotides. STAT6 normally binds to palindromic DNA motifs with a 5'-TTC...GAA-3' core. Notably, the delta-opioid receptor STAT6 site (5'-TTC...GGA-3') is an imperfect palindrome with a mismatch within this core sequence. A systematic analysis of possible mismatch 5'-TTC...GAA-3' motifs revealed that STAT6 also binds to the sequence 5'-TTA...GAA-3'. This motif occurs as a polymorphism in the human mu-opioid receptor gene (Kraus et al. 2001 J. Biol. Chem 276, 43901-43908). We show that this mutated element has a significantly reduced STAT6 binding activity which correlates to its reduced interleukin (IL)-4 inducibility. In contrast, the non-canonical STAT6 site of the delta-opioid receptor binds STAT6 with similar high activity as a perfectly palindromic STAT6 site and is strongly inducible by IL-4.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Base Sequence
- Binding Sites/genetics
- Cell Line, Tumor
- Gene Expression Regulation/drug effects
- Interleukin-4/pharmacology
- Leukocytes/metabolism
- Mice
- Molecular Sequence Data
- Neurons/metabolism
- Polymorphism, Genetic
- Promoter Regions, Genetic/drug effects
- Promoter Regions, Genetic/genetics
- Promoter Regions, Genetic/physiology
- Rats
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/metabolism
- Recombinant Proteins/pharmacology
- STAT6 Transcription Factor
- Trans-Activators/biosynthesis
- Trans-Activators/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Christine Börner
- Department of Pharmacology and Toxicology, University of Magdeburg, Magdeburg, Germany
| | | | | | | |
Collapse
|
45
|
Abstract
Most gut peptides exert their effects through G protein-coupled receptors, a family of about 700 membrane proteins, 87 of which are presently known to have peptide ligands. Three additional gut peptide receptors are not G protein-coupled receptors but regulate intracellular cyclic GMP accumulation. The aim of this review is to illustrate how the sequencing of the human genome and other recent advances in genomics has contributed to our understanding of the role of peptides and their receptors in gastrointestinal function. Recent discoveries include the identification of receptors for the peptides motilin and neuromedin U, and new physiological ligands for the PTH2 receptor, the CRF(2) receptor and the growth hormone secretagogue receptor. Knockout mice lacking specific peptide receptors or their ligands provide informative animal models in which to determine the functions of the numerous peptide-receptor systems in the gut and to predict which of them may be the most fruitful for drug development. Some peptide-receptor signalling systems may be more important in disease states than they are in normal physiology. For example, substance P, galanin, bradykinin and opioids play important roles in visceral pain and inflammation. Other peptides may have developmental roles: for example, disruption of endothelin-3 signalling prevents the normal development of the enteric nervous system and contributes to the pathogenesis of Hirschsprung disease.
Collapse
Affiliation(s)
- Anthony J Harmar
- Division of Neuroscience and Centre for Neuroscience Research, University of Edinburgh, 1 George Square, Edinburgh EH8 9JZ, UK.
| |
Collapse
|
46
|
Börner C, Kraus J, Schröder H, Ammer H, Höllt V. Transcriptional regulation of the human mu-opioid receptor gene by interleukin-6. Mol Pharmacol 2004; 66:1719-26. [PMID: 15448191 DOI: 10.1124/mol.104.003806] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inflammatory pain is counteracted by a number of physiological processes. For example, opioid receptors, which are present on peripheral terminals of sensory neurons, are activated by endogenous opioids, which are released from immune cells migrating to the inflamed tissue. Earlier data demonstrated that interleukin-6 contributes to such inflammation-induced analgesia. In this report, we demonstrated that interleukin-6 strongly induces mu-opioid receptor mRNA in the human neuroblastoma cell line SH SY5Y, whereas delta-opioid receptor mRNA levels are not influenced. The mRNA increase in these cells is followed by an increase in mu-opioid receptor-specific binding. Using transcription factor decoy oligonucleotides, direct evidence was provided that the up-regulation of mu-opioid receptor mRNA in intact cells is dependent on the transcription factors signal transducers and activators of transcription 1 (STAT1) and STAT3, whereas other transcription factors, such as activator protein-1, nuclear factor (NF)-kappaB, or NF-interleukin-6 are not involved. STAT1 and STAT3 bound to a site located at nucleotide -1583 on the promoter of the human mu-opioid receptor gene, as shown by transient transfection experiments, electrophoretic mobility shift assays, and transcription factor decoy oligonucleotides. A mutation analysis of the 5'-TTCATGGAA-3' STAT1/3 element (palindrome underlined) was performed to determine nucleotide residues that are necessary for the binding of STAT1 and STAT3. It suggested that only the palindromic half sides and the two adjacent central nucleotides are required. Neither mutation of the nucleotides outside the palindrome nor mutation of the central nucleotide affected STAT1/3 binding.
Collapse
MESH Headings
- Base Sequence
- Cell Line, Tumor
- DNA Primers
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- Humans
- Inflammation
- Interleukin-6/pharmacology
- Neuroblastoma
- Receptors, Opioid, delta/drug effects
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/genetics
- Receptors, Opioid, mu/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic/drug effects
Collapse
Affiliation(s)
- Christine Börner
- Department of Pharmacology and Toxicology, University of Magdeburg, Magdeburg, Germany
| | | | | | | | | |
Collapse
|
47
|
Greenwood-Van Meerveld B, Gardner CJ, Little PJ, Hicks GA, Dehaven-Hudkins DL. Preclinical studies of opioids and opioid antagonists on gastrointestinal function. Neurogastroenterol Motil 2004; 16 Suppl 2:46-53. [PMID: 15357851 DOI: 10.1111/j.1743-3150.2004.00555.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Opioid receptors in the gastrointestinal (GI) tract mediate the effects of endogenous opioid peptides and exogenously administered opioid analgesics, on a variety of physiological functions associated with motility, secretion and visceral pain. The studies reviewed or reported here describe a range of in vivo activities of opioid receptor antagonists upon GI function in rodents, focusing on mu receptors. Naloxone, and the peripherally acting mu-opioid receptor antagonists alvimopan and methylnaltrexone, reverse morphine-induced inhibition of GI transit in mice and rats, and morphine- or loperamide-induced inhibition of castor oil-induced diarrhoea in mice. At doses producing maximal reversal of morphine-induced effects upon GI transit, only the central nervous system (CNS) penetrant antagonist naloxone was able to reverse morphine-induced analgesia. Both central and peripheral opioid antagonists may affect GI function and/or visceromotor sensitivity in the absence of exogenous opioid analgesics, suggesting a constitutive role for endogenous opioid peptides in the control of GI physiology. Furthermore, in contrast to naloxone, alvimopan does not produce hypersensitivity to the visceromotor response induced by nociceptive levels of colorectal distension in a rodent model of post-inflammatory colonic hypersensitivity, suggesting that in the periphery endogenous mu-opioid receptor-mediated mechanisms do not regulate colonic sensitivity. The data support the hypothesis that peripherally acting opioid antagonists may be able to selectively block opioid receptors in the GI tract, thereby preserving normal GI physiology, while not blocking the effects of endogenous opioid peptides or exogenous opioid analgesics in the CNS. These findings suggest that the primary sites of action of mu-opioid agonists with respect to inhibition of GI function are in the periphery, whereas analgesic activity resides primarily in the CNS.
Collapse
Affiliation(s)
- B Greenwood-Van Meerveld
- Gastrointestinal Research, Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 72104, USA.
| | | | | | | | | |
Collapse
|
48
|
De Schepper HU, Cremonini F, Park MI, Camilleri M. Opioids and the gut: pharmacology and current clinical experience. Neurogastroenterol Motil 2004; 16:383-94. [PMID: 15305992 DOI: 10.1111/j.1365-2982.2004.00513.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This article reviews the pharmacology and physiology of opiate receptors and the current and potential uses of opioid agonists and antagonists in clinical gastroenterology. Mu-receptors are involved in motor and sensory functions, and their modulation is established for treatment of diarrhea. Mu-antagonists have potential to reverse endogenous (e.g., postoperative ileus) or iatrogenic dysmotility (e.g., opioid bowel dysfunction). Modulation of the function of kappa-receptors may be a novel approach to control visceral pain in functional gut disorders. Results of formal testing of novel opioid modulators are keenly awaited.
Collapse
Affiliation(s)
- H U De Schepper
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER) Program, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
49
|
Riazi K, Honar H, Homayoun H, Demehri S, Bahadori M, Dehpour AR. Intestinal inflammation alters the susceptibility to pentylenetetrazole-induced seizure in mice. J Gastroenterol Hepatol 2004; 19:270-7. [PMID: 14748873 DOI: 10.1111/j.1440-1746.2003.03284.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND Inflammatory bowel disorders are associated with increased incidence of seizures. Alteration in the endogenous opioid system and overproduction of nitric oxide have been implicated in colitis. The possible contribution of opioid receptors and nitric oxide to increased seizure susceptibility was examined in a putative model of intestinal inflammation. METHODS The alterations in clonic seizure threshold, induced by pentylenetetrazole, following the induction of intestinal inflammation by the administration of two consecutive daily oral doses of croton oil, was evaluated in mice. This model was used to evaluate the effects of pretreatment with opioid receptor antagonist naltrexone (10 mg/kg, once daily for 4 days or a single dose on the test day), non-specific nitric oxide synthase inhibitor NG-nitro-L-arginine methyl ester (10 mg/kg, once daily), and specific inducible nitric oxide synthase inhibitor aminoguanidine (100 mg/kg, once daily) on seizure threshold in intestinal inflammation. RESULTS A significant decrease in clonic seizure threshold was observed in mice with intestinal inflammation compared to the control group. Chronic pretreatment with naltrexone per se did not cause any significant change in seizure threshold, but it significantly restored the threshold in croton oil-treated mice to that of the control animals. However, acute naltrexone pretreatment (on the test day) could not restore the decreased seizure threshold to control level. Chronic pretreatment with neither NG-nitro-L-arginine methyl ester nor aminoguanidine altered the seizure threshold in the control animals and in mice treated with croton oil. CONCLUSIONS Experimental croton oil-induced intestinal inflammation leads to a proconvulsant effect, which may have clinical relevance. Chronic alterations mediated by endogenous opioids may be involved in this process, though a direct opioid-receptor-mediated effect is unlikely. Nitric oxide synthesis, via constitutive or inducible pathways, is not involved in this increased susceptibility.
Collapse
Affiliation(s)
- Kiarash Riazi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | |
Collapse
|