1
|
Mizrachi T, Marsha O, Brusin K, Ben-David Y, Thakur GA, Vaknin-Dembinsky A, Treinin M, Brenner T. Suppression of neuroinflammation by an allosteric agonist and positive allosteric modulator of the α7 nicotinic acetylcholine receptor GAT107. J Neuroinflammation 2021; 18:99. [PMID: 33902624 PMCID: PMC8077754 DOI: 10.1186/s12974-021-02149-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/03/2021] [Indexed: 02/07/2023] Open
Abstract
Background The α7 nicotinic acetylcholine receptor (α7 nAChR) negatively regulates the synthesis and release of pro-inflammatory cytokines by immune cells. Our previous studies showed that in encephalitogenic T cells, α7 nAChR expression is upregulated and that activation of the cholinergic system can attenuate experimental autoimmune encephalomyelitis (EAE). GAT107 is an allosteric agonist and positive allosteric modulator (ago-PAM) of α7 nAChR that can produce persistent activation of this receptor. Therefore, in the present study, we investigated the effect of GAT107 on neuroinflammation in EAE, the animal model used for the study of multiple sclerosis (MS) via α7 nAChR, and the inflammatory pathways involved. Methods EAE was induced by administration of myelin oligodendrocyte glycoprotein (MOG35–55) in C57BL/6 mice. EAE mice were treated with the ago-PAM GAT107 or a placebo for 9 days, starting from the day of EAE induction. Clinical assessment and immunological evaluation of immune cells and cytokine production was performed. Results Following activation of the α7 nAChR by GAT107 during EAE, disease severity was significantly reduced by 70% and was correlated with a reduction in the extent of neuroinflammation in the CNS. The treatment reduced encephalitogenic T cell proliferation and the production of pro-inflammatory cytokines, as well as increased the production of the anti-inflammatory cytokine IL-10. Furthermore, the expression of immune cell markers was altered by GAT107 treatment, which induced a significant reduction in macrophages, dendritic cells, and B cells, as well as a reduction in anti-MOG35–55 antibodies. Additionally, GAT107 was found to directly activate α7 nAChR in murine macrophage RAW264.7 cells and in human PBMCs derived from MS patients and healthy donors. Conclusions Our results show that GAT107 can be a useful molecule for harnessing the cholinergic anti-inflammatory pathway for long-lasting and wide-ranging modulation and downregulation of neuroinflammation in EAE.
Collapse
Affiliation(s)
- Tehila Mizrachi
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Oshrit Marsha
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Karen Brusin
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Yael Ben-David
- Department of Medical Neurobiology, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ganesh A Thakur
- Pharmaceutical Science, Bouve College of Health Science, Northeastern University, Boston, USA
| | - Adi Vaknin-Dembinsky
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel
| | - Millet Treinin
- Department of Medical Neurobiology, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Talma Brenner
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah University Hospital and Hebrew University Medical School, Jerusalem, Israel.
| |
Collapse
|
2
|
Wu YJ, Wang L, Ji CF, Gu SF, Yin Q, Zuo J. The Role of α7nAChR-Mediated Cholinergic Anti-inflammatory Pathway in Immune Cells. Inflammation 2021; 44:821-834. [PMID: 33405021 DOI: 10.1007/s10753-020-01396-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/05/2020] [Accepted: 12/07/2020] [Indexed: 12/14/2022]
Abstract
Alpha 7 nicotinic acetylcholine receptor (α7nAChR) is widely distributed in the nervous and non-cholinergic immune systems. It is necessary for the cholinergic transmitter to participate in the regulation of inflammatory response and is the key element of cholinergic anti-inflammatory pathway (CAP). Because of the profound impact of CAP on the immune system, α7nAChR is considered as a potential therapeutic target for the treatment of inflammatory diseases. Available evidences confirmed that manipulation of CAP by activating α7nAChR with either endogenous acetylcholine (ACh) or cholinergic agonists can substantially alleviate inflammatory responses both in vivo and in vitro. However, the mechanism through which CAP curbs the excessive pro-inflammatory responses and maintains immune homeostasis is not fully understood. Obtained clues suggest that the crosstalk between CAP and classical inflammatory pathways is the key to elucidate the anti-inflammatory mechanism, and the impacts of CAP activation in α7nAChR-expressing immune cells are the foundation of the immunoregulatory property. In this article, we review and update the knowledge concerning the progresses of α7nAChR-based CAP, including α7nAChR properties, signal transductions, interactions with classic immune pathways, and immunoregulatory functions in different immune cells. Certain critical issues to be addressed are also highlighted. By providing a panoramic view of α7nAChR, the summarized evidences will pave the way for the development of novel anti-inflammatory reagents and strategy and inspire further researches.
Collapse
Affiliation(s)
- Yi-Jin Wu
- The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China
| | - Li Wang
- Department of Pharmacy, Wuhu Medicine and Health School, Wuhu, 241000, China
| | - Chao-Fan Ji
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, China
| | - Shao-Fei Gu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, China
| | - Qin Yin
- The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China.
- School of Pharmacy, Wannan Medical College, Wuhu, 241000, China.
| | - Jian Zuo
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), Wuhu, 241000, China.
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241000, China.
- Research Center of Integrated Traditional and Western Medicine, Wannan Medical College, 241000, Wuhu, China.
| |
Collapse
|
3
|
Polak D, Shmueli A, Brenner T, Shapira L. Oral infection with P. gingivalis exacerbates autoimmune encephalomyelitis. J Periodontol 2018; 89:1461-1466. [PMID: 30030849 DOI: 10.1002/jper.17-0531] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 05/26/2018] [Accepted: 05/27/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Oral infection of mice with P. gingivalis induces periodontal inflammation and attachment loss. The aim of the present study was to investigate whether infection of mice with P. gingivalis, exacerbates the clinical course of experimental autoimmune encephalomyelitis (EAE)-a mouse model of multiple sclerosis (MS). METHODS Induction of EAE was carried out by immunization of C57BL/6 mice with myelin oligodentrocyte glycoprotein (MOG35-55 ). P. gingivalis infection was induced via subcutaneous chambers model and the oral gavage. The severity of EAE was measured using a clinical severity score. Ex-vivo reactivation of lymphocytes with the encephalitogenic peptide MOG35-55 was also tested. RESULTS Subcutaneous as well as oral infection with live P. gingivalis led to significant aggravation of the severity of EAE. Lymph node cells harvested from mice with EAE following P. gingivalis infection showed augmented lymphocyte proliferation towards the encephlatigenic MOG moiety compared to mice with EAE only. CONCLUSIONS The present results indicate that oral infection with P. gingivalis augmented the severity of EAE. This may stem from the systemic pro-inflammatory response triggered by P. gingivalis infection or via antigen mimicking. The present study provides evidence that periodontal infection may play a role as modifier in CNS inflammatory disorders, such as MS.
Collapse
Affiliation(s)
- David Polak
- Department of Periodontology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Aviv Shmueli
- Department of Pediatric Dentistry, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Talma Brenner
- Laboratory of Neuroimmunology, Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lior Shapira
- Department of Periodontology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
4
|
Vakhitova YV, Farafontova EI, Zainullina LF, Vakhitov VA, Tsypysheva IP, Yunusov MS. [Search of (-)-Cytisine Derivatives as Potential Inhibitors of NF-κB and STAT1]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2015; 41:336-45. [PMID: 26502610 DOI: 10.1134/s1068162015030103] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Design and synthesis ofnew derivatives of (-)-cytisine with a wide spectrum of pharmacological activity, represents the potential therapeutic interest for development of drug candidates for neurodegenerative disorders, inflammatory diseases, and treatment of nicotine addiction. We used HEK293 cell line transiently transfected with N F-κB and STATI luciferase reporter constructs to screen the (-)-cytisine derivatives for their potency to modulate basal and induced NF-κB and STAT1 activity. Currently, NF-κB, STAT1 and components of their signaling pathways are considered as attractive targets for pharmacological intervention, primarily in chronic inflammation, cancer, autoimmune, neurodegenerative and infectious diseases. The library of compounds included the derivatives of (-)-cytisine with amino-, amide-, thio- and carboxamide groups at 3, 5 and 12 position of the starting molecule, as well as some bimolecular derivatives. Our experimental data revealed compounds with moderate activating as well as inhibitory effects for basal NF-κB and STATI activity (IC50 or EC50 values are mainly in the micromolar range). The structure-activity relationship analysis demonstrated that the character of activity (activation or inhibition of NFκ-B and STAT1) is determined by the topology of the substituents at the (-)-cytisine molecule, whereas the nature of the substituents mainly contributes to severity of the effect (introduction of aromatic and adamantyl substituents, as well as thionyl or keto groups are of the principal importance). When evaluating the effect of (-)-cytisine derivatives on activity of NF-κB and STATI, induced by specific agents (TNFα and IFNγ, respectively) we observed that some compounds inhibited basal and stimulated activity of NF-κB and STAT1, another compounds showed the dual effect (an increase of basal- and a decrease of stimulated NF-κB activity) and several compounds increase both basal and induced activity of NF-κB and STAT1. Thus, obtained results suggest that one of the possible mechanisms of biological action of (-)-cytisine derivatives is their ability to influence the components of NF-κB and STAT1-dependent signaling pathways.
Collapse
|
5
|
Rouden J, Lasne MC, Blanchet J, Baudoux J. (−)-Cytisine and Derivatives: Synthesis, Reactivity, and Applications. Chem Rev 2013; 114:712-78. [DOI: 10.1021/cr400307e] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Jacques Rouden
- Laboratoire de Chimie Moléculaire
et Thioorganique, ENSICAEN-Université de Caen, CNRS, Institut Normand de Chimie Moléculaire, Médicinale et Macromoléculaire (INC3M), 6 Boulevard du Maréchal Juin, 14050 Caen, France
| | - Marie-Claire Lasne
- Laboratoire de Chimie Moléculaire
et Thioorganique, ENSICAEN-Université de Caen, CNRS, Institut Normand de Chimie Moléculaire, Médicinale et Macromoléculaire (INC3M), 6 Boulevard du Maréchal Juin, 14050 Caen, France
| | - Jérôme Blanchet
- Laboratoire de Chimie Moléculaire
et Thioorganique, ENSICAEN-Université de Caen, CNRS, Institut Normand de Chimie Moléculaire, Médicinale et Macromoléculaire (INC3M), 6 Boulevard du Maréchal Juin, 14050 Caen, France
| | - Jérôme Baudoux
- Laboratoire de Chimie Moléculaire
et Thioorganique, ENSICAEN-Université de Caen, CNRS, Institut Normand de Chimie Moléculaire, Médicinale et Macromoléculaire (INC3M), 6 Boulevard du Maréchal Juin, 14050 Caen, France
| |
Collapse
|
6
|
Ahmed HH, Salem AM, Sabry GM, Husein AA, Kotob SE. Possible therapeutic uses of Salvia triloba and Piper nigrum in Alzheimer's disease-induced rats. J Med Food 2013; 16:437-46. [PMID: 23631499 DOI: 10.1089/jmf.2012.0165] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the role of Salvia triloba L. and Piper nigrum extracts in ameliorating neuroinflammatory insults characteristic of Alzheimer's disease (AD) in an experimentally induced rat model. Adult male Sprague-Dawley rats were classified into Group 1 (n=10): normal healthy animals serving as the negative control group; Group 2 (n=60): the AD-induced group. After AD induction, animals in the AD-induced group were divided randomly and equally into 6 subgroups. The first subgroup served as AD control; the second one, which served as positive control, was treated orally with the conventional therapy for AD (rivastigmine) at a dose of 0.3 mg/kg body weight (b.w.) daily for 3 months. The third and fourth subgroups were, respectively, treated orally with the S. triloba extract at a dose of 750 and 375 mg/kg b.w. daily for 3 months. The fifth and sixth subgroups were, respectively, treated orally with the P. nigrum extract at a dose of 187.5 and 93.75 mg/kg b.w. daily for 3 months. Levels of brain acetylcholine (Ach), serum and brain acetylcholinesterase (AchE) activity, C-reactive protein (CRP), total nuclear factor kappa-B (NF-κB), and monocyte chemoattractant protein-1 (MCP-1) were estimated. The results showed that administration of AlCl3 resulted in a significant elevation in the levels of AchE activity, CRP, NF-κB, and MCP-1 accompanied with a significant depletion in the Ach level. Treatment of AD rats with each of the selected medicinal plant extracts caused marked improvement in the measured biochemical parameters. In conclusion, S. triloba and P. nigrum methanolic extracts have potent anti-inflammatory effects against neuroinflammation characterizing AD.
Collapse
Affiliation(s)
- Hanaa H Ahmed
- Hormones Department, National Research Center, Cairo 12622, Egypt.
| | | | | | | | | |
Collapse
|
7
|
Li YJ, Yang Q, Zhang K, Guo YY, Li XB, Yang L, Zhao MG, Wu YM. Cytisine confers neuronal protection against excitotoxic injury by down-regulating GluN2B-containing NMDA receptors. Neurotoxicology 2013; 34:219-25. [DOI: 10.1016/j.neuro.2012.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 09/08/2012] [Accepted: 09/18/2012] [Indexed: 11/15/2022]
|
8
|
Tasso B, Canu Boido C, Terranova E, Gotti C, Riganti L, Clementi F, Artali R, Bombieri G, Meneghetti F, Sparatore F. Synthesis, Binding, and Modeling Studies of New Cytisine Derivatives, as Ligands for Neuronal Nicotinic Acetylcholine Receptor Subtypes. J Med Chem 2009; 52:4345-57. [DOI: 10.1021/jm900225j] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Bruno Tasso
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Genova, Viale Benedetto XV 3, 16139 Genova, Italy
| | - Caterina Canu Boido
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Genova, Viale Benedetto XV 3, 16139 Genova, Italy
| | - Emanuela Terranova
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Genova, Viale Benedetto XV 3, 16139 Genova, Italy
| | - Cecilia Gotti
- Dipartimento di Farmacologia “E. Trabucchi”, Università degli Studi di Milano, via Vanvitelli 32, 20129 Milano, Italy, Istituto di Neuroscienze del CNR, via Vanvitelli 32, 20129 Milano, Italy
| | - Loredana Riganti
- Dipartimento di Farmacologia “E. Trabucchi”, Università degli Studi di Milano, via Vanvitelli 32, 20129 Milano, Italy, Istituto di Neuroscienze del CNR, via Vanvitelli 32, 20129 Milano, Italy
| | - Francesco Clementi
- Dipartimento di Farmacologia “E. Trabucchi”, Università degli Studi di Milano, via Vanvitelli 32, 20129 Milano, Italy, Istituto di Neuroscienze del CNR, via Vanvitelli 32, 20129 Milano, Italy
| | - Roberto Artali
- Dipartimento di Scienze Farmaceutiche “P. Pratesi”, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Gabriella Bombieri
- Dipartimento di Scienze Farmaceutiche “P. Pratesi”, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Fiorella Meneghetti
- Dipartimento di Scienze Farmaceutiche “P. Pratesi”, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Fabio Sparatore
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Genova, Viale Benedetto XV 3, 16139 Genova, Italy
| |
Collapse
|
9
|
Nizri E, Irony-Tur-Sinai M, Faranesh N, Lavon I, Lavi E, Weinstock M, Brenner T. Suppression of neuroinflammation and immunomodulation by the acetylcholinesterase inhibitor rivastigmine. J Neuroimmunol 2008; 203:12-22. [DOI: 10.1016/j.jneuroim.2008.06.018] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 06/10/2008] [Accepted: 06/10/2008] [Indexed: 11/25/2022]
|
10
|
Brenner T, Nizri E, Irony-Tur-Sinai M, Hamra-Amitay Y, Wirguin I. Acetylcholinesterase inhibitors and cholinergic modulation in Myasthenia Gravis and neuroinflammation. J Neuroimmunol 2008; 201-202:121-7. [DOI: 10.1016/j.jneuroim.2008.05.022] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2008] [Revised: 05/27/2008] [Accepted: 05/28/2008] [Indexed: 10/21/2022]
|