1
|
Carpenter RS, Lagou MK, Karagiannis GS, Maryanovich M. Neural regulation of the thymus: past, current, and future perspectives. Front Immunol 2025; 16:1552979. [PMID: 40046055 PMCID: PMC11880003 DOI: 10.3389/fimmu.2025.1552979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
The thymus is a primary lymphoid organ critical for the development of mature T cells from hematopoietic progenitors. A highly structured organ, the thymus contains distinct regions, precise cytoarchitecture, and molecular signals tightly regulating thymopoiesis. Although the above are well-understood, the structural and functional implications of thymic innervation are largely neglected. In general, neural regulation has become increasingly identified as a critical component of immune cell development and function. The central nervous system (CNS) in the brain coordinates these immunological responses both by direct innervation through peripheral nerves and by neuroendocrine signaling. Yet how these signals, particularly direct neural innervation, may regulate the thymus biology is unclear and understudied. In this review, we highlight historical and current data demonstrating direct neural input to the thymus and assess current evidence of the neural regulation of thymopoiesis. We further discuss the current knowledge gaps and summarize recent advances in techniques that could be used to study how nerves regulate the thymic microenvironment.
Collapse
Affiliation(s)
- Randall S. Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maria K. Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - George S. Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, United States
- Cancer Dormancy Institute, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, United States
- The Marilyn and Stanely M. Katz Institute for Immunotherapy for Cancer and Inflammatory Disorders, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
- Tumor Microenvironment Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
- Cancer Dormancy Institute, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, United States
| |
Collapse
|
2
|
Yang HM, Hou TZ, Zhang YN, Zhao SD, Wu YL, Zhang H. Blocked metabotropic glutamate receptor 5 enhances chemosensitivity in hepatocellular carcinoma and attenuates chemotoxicity in the normal liver by regulating DNA damage. Cancer Gene Ther 2022; 29:1487-1501. [PMID: 35396501 DOI: 10.1038/s41417-022-00465-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
DNA damaging agents are used as chemotherapeutics in many cancers, including hepatocellular carcinoma (HCC). However, they are associated with problems such as low sensitivity to chemotherapy and the induction of liver injury, underscoring the need to identify new therapies. Here, we investigated the differential regulatory effect of metabotropic glutamate receptor 5 (mGlu5) on chemosensitivity in HCC and chemotoxicity to the normal liver. The expression of mGlu5 was higher in HCC than in the normal liver, and correlated with poor prognosis according to The Cancer Genome Atlas database and Integrative Molecular Database of Hepatocellular Carcinoma. Cisplatin, oxaliplatin or methyl methanesulfonate (MMS) caused cell death by decreasing mGlu5 expression in HCC cells and increased mGlu5 expression in hepatic cells. In HCC cells, inhibition of mGlu5 aggravated MMS-induced DNA damage by increasing intracellular Ca2+ overload and mitogen-activated protein kinase (MAPK) activation, thereby promoting cell death, and activation of mGlu5 rescued the effect of MMS. However, in hepatic cells, mGlu5 inhibition alleviated MMS-induced DNA damage by downregulating Ca2+-derived MAPK pathways to advance hepatic cell survival. The opposite effects of mGlu5 overexpression or knockdown on MMS-induced DNA damage supported that cell death is a result of the differential regulation of mGlu5 expression. Inhibition of mGlu5 increased chemosensitivity and decreased chemotoxicity in a rat tumor model. This study suggests that mGlu5 inhibition could act synergistically with HCC chemotherapeutics with minimal side effects, which may improve the treatment of patients with HCC in the future.
Collapse
Affiliation(s)
- Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Tian-Zhong Hou
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Shu-Dong Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, 100083, Beijing, China
| | - Yong-Le Wu
- Center of Hepatic and Digestive Disease, Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
3
|
PEKMEZEKMEK A. MONOSODYUM GLUTAMAT, LEZZET ARTTIRICI MI, ÖLDÜREN LEZZET Mİ? KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1067018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Monosodium glutamat (MSG) 1800’lü yıllardan beri lezzet artırıcı katkı maddesi olarak, işlenmiş ve paketlenmiş tuzlu veya tatlı gıdalarda kullanılmaktadır. Yapılan birçok çalışma MSG kullanımının çok sayıda yapısal ve fonksiyonel bozukluklara yol açabileceğini ortaya çıkarmıştır. Son yıllarda MSG kullanımının çok artması gıda güvenliği konusunda endişelerinde artmasına neden olmuştur.
Collapse
|
4
|
Mprah R, Adzika GK, Gyasi YI, Ndzie Noah ML, Adu-Amankwaah J, Adekunle AO, Duah M, Wowui PI, Weili Q. Glutaminolysis: A Driver of Vascular and Cardiac Remodeling in Pulmonary Arterial Hypertension. Front Cardiovasc Med 2021; 8:667446. [PMID: 33996951 PMCID: PMC8113389 DOI: 10.3389/fcvm.2021.667446] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a decimating ailment described by chronic precapillary pulmonary hypertension, an elevated mean pulmonary arterial pressure with a normal pulmonary capillary wedge pressure, and a raised pulmonary vascular resistance resulting in increased right ventricular afterload culminating in heart failure and death. Current PAH treatments regulate the vasodilatory/vasoconstrictory balance of pulmonary vessels. However, these treatment options are unable to stop the progression of, or reverse, an already established disease. Recent studies have advanced a metabolic dysregulation, featuring increased glutamine metabolism, as a mechanism driving PAH progression. Metabolic dysregulation in PAH leads to increased glutaminolysis to produce substrate to meet the high-energy requirement by hyperproliferative and apoptosis-resistant pulmonary vascular cells. This article explores the role of glutamate metabolism in PAH and how it could be targeted as an anti-remodeling therapeutic strategy.
Collapse
Affiliation(s)
- Richard Mprah
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | | | - Yusif I. Gyasi
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, TX, United States
| | | | | | | | - Maxwell Duah
- Haematology Department, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | | | - Qiao Weili
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
5
|
Anderson G. Tumour Microenvironment: Roles of the Aryl Hydrocarbon Receptor, O-GlcNAcylation, Acetyl-CoA and Melatonergic Pathway in Regulating Dynamic Metabolic Interactions across Cell Types-Tumour Microenvironment and Metabolism. Int J Mol Sci 2020; 22:E141. [PMID: 33375613 PMCID: PMC7795031 DOI: 10.3390/ijms22010141] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/07/2023] Open
Abstract
This article reviews the dynamic interactions of the tumour microenvironment, highlighting the roles of acetyl-CoA and melatonergic pathway regulation in determining the interactions between oxidative phosphorylation (OXPHOS) and glycolysis across the array of cells forming the tumour microenvironment. Many of the factors associated with tumour progression and immune resistance, such as yin yang (YY)1 and glycogen synthase kinase (GSK)3β, regulate acetyl-CoA and the melatonergic pathway, thereby having significant impacts on the dynamic interactions of the different types of cells present in the tumour microenvironment. The association of the aryl hydrocarbon receptor (AhR) with immune suppression in the tumour microenvironment may be mediated by the AhR-induced cytochrome P450 (CYP)1b1-driven 'backward' conversion of melatonin to its immediate precursor N-acetylserotonin (NAS). NAS within tumours and released from tumour microenvironment cells activates the brain-derived neurotrophic factor (BDNF) receptor, TrkB, thereby increasing the survival and proliferation of cancer stem-like cells. Acetyl-CoA is a crucial co-substrate for initiation of the melatonergic pathway, as well as co-ordinating the interactions of OXPHOS and glycolysis in all cells of the tumour microenvironment. This provides a model of the tumour microenvironment that emphasises the roles of acetyl-CoA and the melatonergic pathway in shaping the dynamic intercellular metabolic interactions of the various cells within the tumour microenvironment. The potentiation of YY1 and GSK3β by O-GlcNAcylation will drive changes in metabolism in tumours and tumour microenvironment cells in association with their regulation of the melatonergic pathway. The emphasis on metabolic interactions across cell types in the tumour microenvironment provides novel future research and treatment directions.
Collapse
Affiliation(s)
- George Anderson
- Clinical Research Communications (CRC) Scotland & London, Eccleston Square, London SW1V 6UT, UK
| |
Collapse
|
6
|
Behavioral responses of mGluR3-KO mice to the lipopolysaccharide-induced innate inflammatory reaction. Pharmacol Biochem Behav 2020; 190:172852. [DOI: 10.1016/j.pbb.2020.172852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 12/13/2022]
|
7
|
Sadat-Shirazi MS, Vousooghi N, Alizadeh B, Makki SM, Zarei SZ, Nazari S, Zarrindast MR. Expression of NMDA receptor subunits in human blood lymphocytes: A peripheral biomarker in online computer game addiction. J Behav Addict 2018; 7:260-268. [PMID: 29788757 PMCID: PMC6174581 DOI: 10.1556/2006.7.2018.35] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background and aims Repeated performance of some behaviors such as playing computer games could result in addiction. The NMDA receptor is critically involved in the development of behavioral and drug addictions. It has been claimed that the expression level of neurotransmitter receptors in the brain may be reflected in peripheral blood lymphocytes (PBLs). Methods Here, using a real-time PCR method, we have investigated the mRNA expression of GluN2A, GluN2D, GluN3A, and GluN3B subunits of the NMDA receptor in PBLs of male online computer game addicts (n = 25) in comparison with normal subjects (n = 26). Results Expression levels of GluN2A, GluN2D, and GluN3B subunits were not statistically different between game addicts and the control group. However, the mRNA expression of the GluN3A subunit was downregulated in PBLs of game addicts. Discussion and conclusions Transcriptional levels of GluN2A and GluN2D subunits in online computer game addicts are similar to our previously reported data of opioid addiction and are not different from the control group. However, unlike our earlier finding of drug addiction, the mRNA expression levels of GluN3A and GluN3B subunits in PBLs of game addicts are reduced and unchanged, respectively, compared with control subjects. It seems that the downregulated state of the GluN3A subunit of NMDA receptor in online computer game addicts is a finding that deserves more studies in the future to see whether it can serve as a peripheral biomarker in addiction studies, where the researcher wants to rule out the confusing effects of abused drugs.
Collapse
Affiliation(s)
- Mitra-Sadat Sadat-Shirazi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Vousooghi
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran,Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran,Corresponding author: Nasim Vousooghi, Pharm D, PhD; Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, PO Box 1417755469, Tehran, Iran; Phone: +98 21 8899 1118; Fax: +98 21 8899 1117; E-mail:
| | - Bentolhoda Alizadeh
- Department of Biology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Seyed Mohammad Makki
- Department of Psychiatry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Shahrzad Nazari
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zarrindast
- Genetics Laboratory, Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,School of Cognitive Sciences, Institute for Studies in Theoretical Physics and Mathematics, Tehran, Iran,Institute for Cognitive Science Studies, Tehran, Iran
| |
Collapse
|
8
|
Fazio F, Ulivieri M, Volpi C, Gargaro M, Fallarino F. Targeting metabotropic glutamate receptors for the treatment of neuroinflammation. Curr Opin Pharmacol 2018; 38:16-23. [PMID: 29471184 DOI: 10.1016/j.coph.2018.01.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/31/2018] [Indexed: 12/15/2022]
Abstract
A large body of evidence suggests that neuroinflammation lies at the core of nearly all CNS disorders, including psychiatric disorders. Invading and local immune cells orchestrate the series of events that lead to either tissue repair or damage in response to neuroinflammation. Both lymphocytes and microglia express metabotropic glutamate (mGlu) receptors, which respond to glutamate or other endogenous activators (e.g. some kynurenine metabolites of tryptophan metabolism) influencing immune phenotype and the balance between pro-inflammatory and anti-inflammatory cytokines. Here, we offer an up-to-date on the role of individual mGlu receptor subtypes in the regulation of innate and adaptive immune response, highlighting the relevance of this information in the development of subtype-selective mGlu receptor ligands for treatment of CNS disorders.
Collapse
Affiliation(s)
| | - Martina Ulivieri
- Department of Physiology and Pharmacology, Sapienza University, Rome, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| | - Francesca Fallarino
- Department of Experimental Medicine, University of Perugia, Polo Unico Sant'Andrea delle Fratte, Piazzale Gambuli, 06132 Perugia, Italy
| |
Collapse
|
9
|
Ferrigno A, Berardo C, Di Pasqua LG, Siciliano V, Richelmi P, Nicoletti F, Vairetti M. Selective Blockade of the Metabotropic Glutamate Receptor mGluR5 Protects Mouse Livers in In Vitro and Ex Vivo Models of Ischemia Reperfusion Injury. Int J Mol Sci 2018; 19:E314. [PMID: 29360756 PMCID: PMC5855547 DOI: 10.3390/ijms19020314] [Citation(s) in RCA: 12] [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: 11/20/2017] [Revised: 01/18/2018] [Accepted: 01/22/2018] [Indexed: 12/13/2022] Open
Abstract
2-Methyl-6-(phenylethynyl)pyridine (MPEP), a negative allosteric modulator of the metabotropic glutamate receptor (mGluR) 5, protects hepatocytes from ischemic injury. In astrocytes and microglia, MPEP depletes ATP. These findings seem to be self-contradictory, since ATP depletion is a fundamental stressor in ischemia. This study attempted to reconstruct the mechanism of MPEP-mediated ATP depletion and the consequences of ATP depletion on protection against ischemic injury. We compared the effects of MPEP and other mGluR5 negative modulators on ATP concentration when measured in rat hepatocytes and acellular solutions. We also evaluated the effects of mGluR5 blockade on viability in rat hepatocytes exposed to hypoxia. Furthermore, we studied the effects of MPEP treatment on mouse livers subjected to cold ischemia and warm ischemia reperfusion. We found that MPEP and 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl]pyridine (MTEP) deplete ATP in hepatocytes and acellular solutions, unlike fenobam. This finding suggests that mGluR5s may not be involved, contrary to previous reports. MPEP, as well as MTEP and fenobam, improved hypoxic hepatocyte viability, suggesting that protection against ischemic injury is independent of ATP depletion. Significantly, MPEP protected mouse livers in two different ex vivo models of ischemia reperfusion injury, suggesting its possible protective deployment in the treatment of hepatic inflammatory conditions.
Collapse
Affiliation(s)
- Andrea Ferrigno
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| | - Clarissa Berardo
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| | - Laura Giuseppina Di Pasqua
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| | - Veronica Siciliano
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| | - Plinio Richelmi
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, Sapienza University, 00185 Roma, Italy.
- I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy.
| | - Mariapia Vairetti
- Department of Internal Medicine and Therapeutics, Cellular and Molecular Pharmacology and Toxicology Unit, University of Pavia, 27100 Pavia, Italy.
| |
Collapse
|
10
|
Metabotropic glutamate receptor 5 deficiency inhibits neutrophil infiltration after traumatic brain injury in mice. Sci Rep 2017; 7:9998. [PMID: 28855570 PMCID: PMC5577182 DOI: 10.1038/s41598-017-10201-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 08/07/2017] [Indexed: 11/08/2022] Open
Abstract
Both brain native inflammatory cells and infiltrated peripheral white blood cells (WBCs) are primary participants in the brain inflammatory damage post-TBI. Metabotropic glutamate receptor 5 (mGluR5) has been reported to regulate microglias and astrocytes to affect inflammation after TBI, but its effect on modulating infiltrated peripheral WBCs remains unclear. In a mouse moderate TBI model, we found that mGluR5 knockout (KO) significantly reduced neutrophil infiltration and inflammatory cytokine expression in the brain at 24 hours post TBI, which was accompanied by improved neurological dysfunction. Further investigation indicated that mGluR5 KO reduced the permeability of blood-brain barrier (BBB), the entrance for neutrophils to enter brain, and markedly decreased the mRNA levels of neutrophil-associated chemokines in brain tissue, including CXCL1, CXCL2, CCL2, CCL4 and CCL5. Using brain microvascular endothelial cells (BMECs), neutrophils and a BBB model in vitro, we confirmed the inhibitory effect of mGluR5 deficiency on neutrophil infiltration and demonstrated that blockade of protein kinase C (PKC) signaling was involved in it. These results provide insight into the role of mGluR5 in the regulation of inflammation in the acute phase of TBI, which may provide novel clues for TBI therapy.
Collapse
|
11
|
Ferrigno A, Berardo C, Di Pasqua LG, Siciliano V, Richelmi P, Vairetti M. Localization and role of metabotropic glutamate receptors subtype 5 in the gastrointestinal tract. World J Gastroenterol 2017; 23:4500-4507. [PMID: 28740338 PMCID: PMC5504365 DOI: 10.3748/wjg.v23.i25.4500] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 05/03/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023] Open
Abstract
Metabotropic glutamate receptor subtype 5 (mGluR5) is a Group I mGlu subfamily of receptors coupled to the inositol trisphosphate/diacylglycerol pathway. Like other mGluR subtypes, mGluR5s contain a phylogenetically conserved, extracellular orthosteric binding site and a more variable allosteric binding site, located on the heptahelical transmembrane domain. The mGluR5 receptor has proved to be a key pharmacological target in conditions affecting the central nervous system (CNS) but its presence outside the CNS underscores its potential role in pathologies affecting peripheral organs such as the gastrointestinal (GI) tract and accessory digestive organs such as the tongue, liver and pancreas. Following identification of mGluR5s in the mouth, various studies have subsequently demonstrated its involvement in mechanical allodynia, inflammation, pain and oral cancer. mGluR5 expression has also been identified in gastroesophageal vagal pathways. Indeed, experimental and human studies have demonstrated that mGluR5 blockade reduces transient lower sphincter relaxation and reflux episodes. In the intestine, mGluR5s have been shown to be involved in the control of intestinal inflammation, visceral pain and the epithelial barrier function. In the liver, mGluR5s have a permissive role in the onset of ischemic injury in rat and mice hepatocytes. Conversely, livers from mice treated with selective negative allosteric modulators and mGluR5 knockout mice are protected against ischemic injury. Similar results have been observed in experimental models of free-radical injury and in vivo mouse models of acetaminophen intoxication. Finally, mGluR5s in the pancreas are associated with insulin secretion control. The picture is, however, far from complete as the review attempts to establish in particular as regards identifying specific targets and innovative therapeutic approaches for the treatment of GI disorders.
Collapse
|
12
|
Levite M. Glutamate, T cells and multiple sclerosis. J Neural Transm (Vienna) 2017; 124:775-798. [PMID: 28236206 DOI: 10.1007/s00702-016-1661-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 11/25/2016] [Indexed: 12/18/2022]
Abstract
Glutamate is the major excitatory neurotransmitter in the nervous system, where it induces multiple beneficial and essential effects. Yet, excess glutamate, evident in a kaleidoscope of acute and chronic pathologies, is absolutely catastrophic, since it induces excitotoxicity and massive loss of brain function. Both the beneficial and the detrimental effects of glutamate are mediated by a large family of glutamate receptors (GluRs): the ionotropic glutamate receptors (iGluRs) and the metabotropic glutamate receptors (mGluRs), expressed by most/all cells of the nervous system, and also by many non-neural cells in various peripheral organs and tissues. T cells express on their cell surface several types of functional GluRs, and so do few other immune cells. Furthermore, glutamate by itself activates resting normal human T cells, and induces/elevates key T cell functions, among them: T cell adhesion, chemotactic migration, cytokine secretion, gene expression and more. Glutamate has also potent effects on antigen/mitogen/cytokine-activated T cells. Furthermore, T cells can even produce and release glutamate, and affect other cells and themselves via their own glutamate. Multiple sclerosis (MS) and its animal model Experimental Autoimmune Encephalomyelitis (EAE) are mediated by autoimmune T cells. In MS and EAE, there are excess glutamate levels, and multiple abnormalities in glutamate degrading enzymes, glutamate transporters, glutamate receptors and glutamate signaling. Some GluR antagonists block EAE. Enhancer of mGluR4 protects from EAE via regulatory T cells (Tregs), while mGluR4 deficiency exacerbates EAE. The protective effect of mGluR4 on EAE calls for testing GluR4 enhancers in MS patients. Oral MS therapeutics, namely Fingolimod, dimethyl fumarate and their respective metabolites Fingolimod-phosphate and monomethyl fumarate, can protect neurons against acute glutamatergic excitotoxic damage. Furthermore, Fingolimod reduce glutamate-mediated intracortical excitability in relapsing-remitting MS. Glatiramer acetate -COPAXONE®, an immunomodulator drug for MS, reverses TNF-α-induced alterations of striatal glutamate-mediated excitatory postsynaptic currents in EAE-afflicted mice. With regard to T cells of MS patients: (1) The cell surface expression of a specific GluR: the AMPA GluR3 is elevated in T cells of MS patients during relapse and with active disease, (2) Glutamate and AMPA (a selective agonist for glutamate/AMPA iGluRs) augment chemotactic migration of T cells of MS patients, (3) Glutamate augments proliferation of T cells of MS patients in response to myelin-derived proteins: MBP and MOG, (4) T cells of MS patients respond abnormally to glutamate, (5) Significantly higher proliferation values in response to glutamate were found in MS patients assessed during relapse, and in those with gadolinium (Gd)+ enhancing lesions on MRI. Furthermore, glutamate released from autoreactive T cells induces excitotoxic cell death of neurons. Taken together, the evidences accumulated thus far indicate that abnormal glutamate levels and signaling in the nervous system, direct activation of T cells by glutamate, and glutamate release by T cells, can all contribute to MS. This may be true also to other neurological diseases. It is postulated herein that the detrimental activation of autoimmune T cells by glutamate in MS could lead to: (1) Cytotoxicity in the CNS: T cell-mediated killing of neurons and glia cells, which would subsequently increase the extracellular glutamate levels, and by doing so increase the excitotoxicity mediated by excess glutamate, (2) Release of proinflammatory cytokines, e.g., TNFα and IFNγ that increase neuroinflammation. Finally, if excess glutamate, abnormal neuronal signaling, glutamate-induced activation of T cells, and glutamate release by T cells are indeed all playing a key detrimental role in MS, then optional therapeutic tolls include GluR antagonists, although these may have various side effects. In addition, an especially attractive therapeutic strategy is the novel and entirely different therapeutic approach to minimize excess glutamate and excitotoxicity, titled: 'brain to blood glutamate scavenging', designed to lower excess glutamate levels in the CNS by 'pumping it out' from the brain to the blood. The glutamate scavanging is achieved by lowering glutamate levels in the blood by intravenous injection of the blood enzyme glutamate oxaloacetate transaminase (GOT). The glutamate-scavenging technology, which is still experimental, validated so far for other brain pathologies, but not tested on MS or EAE yet, may be beneficial for MS too, since it could decrease both the deleterious effects of excess glutamate on neural cells, and the activation of autoimmune T cells by glutamate in the brain. The topic of glutamate scavenging, and also its potential benefit for MS, are discussed towards the end of the review, and call for research in this direction.
Collapse
Affiliation(s)
- Mia Levite
- Faculty of Medicine, School of Pharmacy, The Hebrew University, Jerusalem, Israel. .,Institute of Gene Therapy, Hadassah Medical Center, 91120, Ein Karem, Jerusalem, Israel.
| |
Collapse
|
13
|
Rees CL, White CM, Ascoli GA. Neurochemical Markers in the Mammalian Brain: Structure, Roles in Synaptic Communication, and Pharmacological Relevance. Curr Med Chem 2017; 24:3077-3103. [PMID: 28413962 PMCID: PMC5646670 DOI: 10.2174/0929867324666170414163506] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/15/2017] [Accepted: 04/10/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND Knowledge of molecular marker (typically protein or mRNA) expression in neural systems can provide insight to the chemical blueprint of signal processing and transmission, assist in tracking developmental or pathological progressions, and yield key information regarding potential medicinal targets. These markers are particularly relevant in the mammalian brain in the light of its unsurpassed cellular diversity. Accordingly, molecular expression profiling is rapidly becoming a major approach to classify neuron types. Despite a profusion of research, however, the biological functions of molecular markers commonly used to distinguish neuron types remain incompletely understood. Furthermore, most molecular markers of mammalian neuron types are also present in other organs, therefore complicating considerations of their potential pharmacological interactions. OBJECTIVE Here, we survey 15 prominent neurochemical markers from five categories, namely membrane transporters, calcium-binding proteins, neuropeptides, receptors, and extracellular matrix proteins, explaining their relation and relevance to synaptic communication. METHOD For each marker, we summarize fundamental structural features, cellular functionality, distributions within and outside the brain, as well as known drug effectors and mechanisms of action. CONCLUSION This essential primer thus links together the cellular complexity of the brain, the chemical properties of key molecular players in neurotransmission, and possible biomedical opportunities.
Collapse
Affiliation(s)
- Christopher L. Rees
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Charise M. White
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| | - Giorgio A. Ascoli
- Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA, USA
| |
Collapse
|
14
|
Peterlik D, Stangl C, Bauer A, Bludau A, Keller J, Grabski D, Killian T, Schmidt D, Zajicek F, Jaeschke G, Lindemann L, Reber SO, Flor PJ, Uschold-Schmidt N. Blocking metabotropic glutamate receptor subtype 5 relieves maladaptive chronic stress consequences. Brain Behav Immun 2017; 59:79-92. [PMID: 27524668 DOI: 10.1016/j.bbi.2016.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/29/2016] [Accepted: 08/10/2016] [Indexed: 12/22/2022] Open
Abstract
Etiology and pharmacotherapy of stress-related psychiatric conditions and somatoform disorders are areas of high unmet medical need. Stressors holding chronic plus psychosocial components thereby bear the highest health risk. Although the metabotropic glutamate receptor subtype 5 (mGlu5) is well studied in the context of acute stress-induced behaviors and physiology, virtually nothing is known about its potential involvement in chronic psychosocial stress. Using the mGlu5 negative allosteric modulator CTEP (2-chloro-4-[2-[2,5-dimethyl-1-[4-(trifluoromethoxy)phenyl]imidazol-4yl]ethynyl]pyridine), a close analogue of the clinically active drug basimglurant - but optimized for rodent studies, as well as mGlu5-deficient mice in combination with a mouse model of male subordination (termed CSC, chronic subordinate colony housing), we demonstrate that mGlu5 mediates multiple physiological, immunological, and behavioral consequences of chronic psychosocial stressor exposure. For instance, CTEP dose-dependently relieved hypothalamo-pituitary-adrenal axis dysfunctions, colonic inflammation as well as the CSC-induced increase in innate anxiety; genetic ablation of mGlu5 in mice largely reproduced the stress-protective effects of CTEP and additionally ameliorated CSC-induced physiological anxiety. Interestingly, CSC also induced an upregulation of mGlu5 in the hippocampus, a stress-regulating brain area. Taken together, our findings provide evidence that mGlu5 is an important mediator for a wide range of chronic psychosocial stress-induced alterations and a potentially valuable drug target for the treatment of chronic stress-related pathologies in man.
Collapse
Affiliation(s)
- Daniel Peterlik
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Christina Stangl
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Amelie Bauer
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Anna Bludau
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Jana Keller
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Dominik Grabski
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Tobias Killian
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Dominic Schmidt
- Institute of Immunology, University of Regensburg, D-93042 Regensburg, Germany
| | - Franziska Zajicek
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany
| | - Georg Jaeschke
- Roche Pharmaceutical Research and Early Development, Discovery Chemistry, Roche Innovation Center Basel, CH-4070 Basel, Switzerland
| | - Lothar Lindemann
- Roche Pharmaceutical Research and Early Development, Discovery Neuroscience, Neuroscience, Ophthalmology, and Rare Diseases, Roche Innovation Center Basel, CH-4070 Basel, Switzerland
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Clinic for Psychosomatic Medicine and Psychotherapy, University of Ulm, D-89081 Ulm, Germany
| | - Peter J Flor
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany.
| | - Nicole Uschold-Schmidt
- Faculty of Biology and Preclinical Medicine, Laboratory of Molecular and Cellular Neurobiology, University of Regensburg, D-93053 Regensburg, Germany.
| |
Collapse
|
15
|
Ferrigno A, Vairetti M, Ambrosi G, Rizzo V, Richelmi P, Blandini F, Fuzzati-Armentero MT. Selective blockade of mGlu5 metabotropic glutamate receptors is protective against hepatic mitochondrial dysfunction in 6-OHDA lesioned Parkinsonian rats. Clin Exp Pharmacol Physiol 2016; 42:695-703. [PMID: 25904005 DOI: 10.1111/1440-1681.12410] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 12/21/2022]
Abstract
Non-motor symptoms including those involving the splanchnic district are present in Parkinson's disease (PD). The authors previously reported that PD-like rats, bearing a lesion of the nigrostriatal pathway induced by the injection of 6-hydroxydopamine (6-OHDA), have impaired hepatic mitochondrial function. Glutamate intervenes at multiple levels in PD and liver pathophysiologies. The metabotropic glutamate receptor 5 (mGluR5) is abundantly expressed in brain and liver and may represent a pharmacological target for PD therapy. This study investigated whether and how chronic treatment with 2-methyl-6-(phenylethynyl)-pyridine (MPEP), a well-characterized mGluR5 antagonist, may influence hepatic function with regard to neuronal cell loss in PD-like rats. Chronic treatment with MPEP was started immediately (Early) or 4 weeks after (Delayed) intrastriatal injection of 6-OHDA and lasted 4 weeks. Early MPEP treatment significantly prevented the decrease in adenosine triphosphate (ATP) production/content and counteracted increased reactive oxygen species (ROS) formation in isolated hepatic mitochondria of PD-like animals. Early MPEP administration also reduced the toxin-induced neurodegenerative process; improved survival of nigral dopaminergic neurons correlated with enhanced mitochondrial ATP content and production. ATP content/production, in turn, negatively correlated with ROS formation suggesting that the MPEP-dependent improvement in hepatic function positively influenced neuronal cell survival. Delayed MPEP treatment had no effect on hepatic mitochondrial function and neuronal cell loss. Antagonizing mGluR5 may synergistically act against neuronal cell loss and PD-related hepatic mitochondrial alterations and may represent an interesting alternative to non-dopaminergic therapeutic strategies for the treatment of PD.
Collapse
Affiliation(s)
- Andrea Ferrigno
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Mariapia Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Giulia Ambrosi
- Center for Research in Neurodegenerative Diseases, Laboratory of Functional Neurochemistry, National Neurological Institute C. Mondino, Pavia, Italy
| | - Vittoria Rizzo
- Department of Molecular Medicine, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Plinio Richelmi
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Fabio Blandini
- Center for Research in Neurodegenerative Diseases, Laboratory of Functional Neurochemistry, National Neurological Institute C. Mondino, Pavia, Italy
| | - Marie-Therese Fuzzati-Armentero
- Center for Research in Neurodegenerative Diseases, Laboratory of Functional Neurochemistry, National Neurological Institute C. Mondino, Pavia, Italy
| |
Collapse
|
16
|
Medial Septal NMDA Glutamate Receptors are Involved in Modulation of Blood Natural Killer Cell Activity in Rats. J Neuroimmune Pharmacol 2015; 11:121-32. [PMID: 26454750 DOI: 10.1007/s11481-015-9632-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 09/15/2015] [Indexed: 02/08/2023]
Abstract
The purpose of the present study was to determine the specific role of the medial septal (MS) NMDA glutamate receptors on peripheral blood natural killer cell cytotoxicity (NKCC) and their (large granular lymphocyte, LGL) number, as well as the plasma concentration of tumor necrosis factor α (TNF-α) and corticosterone in male Wistar rats exposed to elevated plus maze (EPM) stress or non-stress conditions. The NMDA groups were injected with NMDA glutamate receptor agonist (N-methyl-D-aspartate; 0.25 μg/rat), the D-AP7 group was injected with DL-2-amino-7-phosphoheptanoate (0.1 μg/rat), an antagonist of NMDA glutamate receptors, and the control Sal group with saline (0.5 μl/rat) via previously implanted cannulae into the MS. There was an increase in the NKCC, NK/LGL number and plasma TNF-α concentration after the NMDA injections, being much stronger within the rats under non-stress conditions rather than the rats exposed to EPM stress. These parameters were decreased in the D-AP7 rats, suggesting receptor/ion channel specificity. Moreover, a lower plasma corticosterone concentration within the NMDA rather than the Sal and D-AP7 groups was found. The obtained results suggest that activation of the NMDA glutamate receptors in the MS, accompanied by changes in the corticosterone and cytokine responses, may be involved in modulation of the blood natural anti-tumor response, under EPM stress and non-stress conditions.
Collapse
|
17
|
Gupta R, Palchaudhuri S, Chattopadhyay D. Glutamate induces neutrophil cell migration by activating class I metabotropic glutamate receptors. Amino Acids 2012; 44:757-67. [DOI: 10.1007/s00726-012-1400-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 09/05/2012] [Indexed: 12/11/2022]
|
18
|
Targeting metabotropic glutamate receptors in neuroimmune communication. Neuropharmacology 2012; 63:501-6. [PMID: 22640632 DOI: 10.1016/j.neuropharm.2012.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/15/2012] [Accepted: 05/17/2012] [Indexed: 01/13/2023]
Abstract
L-Glutamate (L-Glu) is the principal excitatory neurotransmitter in the Central Nervous System (CNS), where it regulates cellular and synaptic activity, neuronal plasticity, cell survival and other relevant functions. Glutamatergic neurotransmission is complex and involves both ionotropic (ligand-gated ion channels; iGluRs) and metabotropic receptors (G-protein coupled receptors). Recent evidence suggests that glutamatergic receptors are also expressed by immune cells, regulating the degree of cell activation. In this review we primarily focus on mGluRs and their role in the crosstalk between the central nervous and immune systems during neuroinflammation.
Collapse
|
19
|
Boldyrev AA, Bryushkova EA, Vladychenskaya EA. NMDA receptors in immune competent cells. BIOCHEMISTRY (MOSCOW) 2012; 77:128-34. [DOI: 10.1134/s0006297912020022] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
20
|
Julio-Pieper M, Flor PJ, Dinan TG, Cryan JF. Exciting times beyond the brain: metabotropic glutamate receptors in peripheral and non-neural tissues. Pharmacol Rev 2011; 63:35-58. [PMID: 21228260 DOI: 10.1124/pr.110.004036] [Citation(s) in RCA: 146] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Metabotropic glutamate (mGlu) receptors are G-protein-coupled receptors expressed primarily on neurons and glial cells, where they are located in the proximity of the synaptic cleft. In the central nervous system (CNS), mGlu receptors modulate the effects of l-glutamate neurotransmission in addition to that of a variety of other neurotransmitters. However, mGlu receptors also have a widespread distribution outside the CNS that has been somewhat neglected to date. Based on this expression, diverse roles of mGlu receptors have been suggested in a variety of processes in health and disease including controlling hormone production in the adrenal gland and pancreas, regulating mineralization in the developing cartilage, modulating lymphocyte cytokine production, directing the state of differentiation in embryonic stem cells, and modulating gastrointestinal secretory function. Understanding the role of mGlu receptors in the periphery will also provide a better insight into potential side effects of drugs currently being developed for neurological and psychiatric conditions. This review summarizes the new potential roles of mGlu receptors and raises the possibility of novel pharmacological targets for various disorders.
Collapse
Affiliation(s)
- Marcela Julio-Pieper
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
21
|
Haas HS, Linecker A, Pfragner R, Sadjak A. Peripheral glutamate signaling in head and neck areas. Head Neck 2011; 32:1554-72. [PMID: 20848447 DOI: 10.1002/hed.21438] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The major excitatory neurotransmitter glutamate is also found in the periphery in an increasing number of nonexcitable cells. In line with this it became apparent that glutamate can regulate a broad array of peripheral biological responses, as well. Of particular interest is the discovery that glutamate receptor reactive reagents can influence tumor biology. However, the knowledge of glutamate signaling in peripheral tissues is still incomplete and, in the case of head and neck areas, is almost lacking. The roles of glutamate signaling pathways in these regions are manifold and include orofacial pain, periodontal bone production, skin and airway inflammation, as well as salivation. Furthermore, the interrelations between glutamate and cancers in the oral cavity, thyroid gland, and other regions are discussed. In summary, this review shall strengthen the view that glutamate receptor reagents may also be promising targets for novel therapeutic concepts suitable for a number of diseases in peripheral tissues. The contents of this review cover the following sections: Introduction; The "Glutamate System"; The Taste of Glutamate; Glutamate Signaling in Dental Regions; Glutamate Signaling in Head and Neck Areas; Glutamate Signaling in Head and Neck Cancer; A Brief Overview of Glutamate Signaling in Other Cancers; and Conclusion.
Collapse
Affiliation(s)
- Helga Susanne Haas
- Department of Pathophysiology and Immunology, Center of Molecular Medicine, Medical University of Graz, Graz, Austria.
| | | | | | | |
Collapse
|
22
|
Affaticati P, Mignen O, Jambou F, Potier MC, Klingel-Schmitt I, Degrouard J, Peineau S, Gouadon E, Collingridge GL, Liblau R, Capiod T, Cohen-Kaminsky S. Sustained calcium signalling and caspase-3 activation involve NMDA receptors in thymocytes in contact with dendritic cells. Cell Death Differ 2011; 18:99-108. [PMID: 20577261 PMCID: PMC3131881 DOI: 10.1038/cdd.2010.79] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 04/27/2010] [Accepted: 05/17/2010] [Indexed: 12/15/2022] Open
Abstract
L-glutamate, the major excitatory neurotransmitter, also has a role in non-neuronal tissues and modulates immune responses. Whether NMDA receptor (NMDAR) signalling is involved in T-cell development is unknown. In this study, we show that mouse thymocytes expressed an array of glutamate receptors, including NMDARs subunits. Sustained calcium (Ca(2+)) signals and caspase-3 activation in thymocytes were induced by interaction with antigen-pulsed dendritic cells (DCs) and were inhibited by NMDAR antagonists MK801 and memantine. NMDARs were transiently activated, triggered the sustained Ca(2+) signal and were corecruited with the PDZ-domain adaptor postsynaptic density (PSD)-95 to thymocyte-DC contact zones. Although T-cell receptor (TCR) activation was sufficient for relocalization of NMDAR and PSD-95 at the contact zone, NMDAR could be activated only in a synaptic context. In these T-DC contacts, thymocyte activation occurred in the absence of exogenous glutamate, indicating that DCs could be a physiological source of glutamate. DCs expressed glutamate, glutamate-specific vesicular glutamate transporters and were capable of fast glutamate release through a Ca(2+)-dependent mechanism. We suggest that glutamate released by DCs could elicit focal responses through NMDAR-signalling in T cells undergoing apoptosis. Thus, synapses between T and DCs could provide a functional platform for coupling TCR activation and NMDAR signalling, which might reflect on T-cell development and modulation of the immune response.
Collapse
Affiliation(s)
- P Affaticati
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
| | - O Mignen
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
| | - F Jambou
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
| | - M-C Potier
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche UMR 7637, Département de Neurobiologie et Diversité Cellulaire, ESPCI, Paris F-75005, France
| | - I Klingel-Schmitt
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
- Institut National de la Recherche Médicale, INSERM U999, Institut d'Immunologie, Hématologie, Pneumologie, Le Plessis Robinson F-92350, France
| | - J Degrouard
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche UMR 8080, Département de Microscopie Electronique, Université Paris-Sud, Orsay F-91405, France
| | - S Peineau
- Department of Anatomy, MRC Centre for Synaptic Plasticity, School of Medical Sciences, Bristol, UK
- Institut National de la Recherche Médicale, INSERM, U676, Département de physiopathologie et neuroprotection des atteintes du cerveau en développement, Hôpital Robert Debré, Université Paris 7, Faculté de Médecine Denis Diderot Paris, Paris F-75019, France
| | - E Gouadon
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
- Institut National de la Recherche Médicale, INSERM U999, Institut d'Immunologie, Hématologie, Pneumologie, Le Plessis Robinson F-92350, France
| | - G L Collingridge
- Department of Anatomy, MRC Centre for Synaptic Plasticity, School of Medical Sciences, Bristol, UK
- Department of Psychiatry, Brain Research Centre, University of British Columbia, Vancouver, British Columbia V6T-1Z3, Canada
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul 151747, Korea
| | - R Liblau
- Institut National de la Santé et de la Recherche Médicale, INSERM U563, Département d'Immunologie et Maladies Infectieuses, Hôpital Universitaire Purpan, Toulouse, F-31000, France
| | - T Capiod
- Institut National de la Santé et de la Recherche Médicale, INSERM, U1003, IFR147, Université Lille 1, Villeneuve d'Ascq F-59655, France
| | - S Cohen-Kaminsky
- Centre National de la Recherche Scientifique (CNRS), Département des Sciences du Vivant (DSV), Institut National des Sciences Biologiques (INSB), Unité Mixte de Recherche UMR 8162, Institut Paris Sud Cytokines, Le Plessis Robinson, Ile de France F-92350, France
- Univ. Paris-Sud, Orsay F-91405, Faculté de Médecine Paris-Sud, Le Kremlin-Bicêtre F-94270, France
- Hôpital Marie Lannelongue, Département de Recherche Médicale, Le Plessis-Robinson F-92350, France
- Institut National de la Recherche Médicale, INSERM U999, Institut d'Immunologie, Hématologie, Pneumologie, Le Plessis Robinson F-92350, France
| |
Collapse
|
23
|
Kim JH, Lee JO, Lee SK, Moon JW, You GY, Kim SJ, Park SH, Park JM, Lim SY, Suh PG, Uhm KO, Song MS, Kim HS. The glutamate agonist homocysteine sulfinic acid stimulates glucose uptake through the calcium-dependent AMPK-p38 MAPK-protein kinase C zeta pathway in skeletal muscle cells. J Biol Chem 2010; 286:7567-76. [PMID: 21193401 DOI: 10.1074/jbc.m110.149328] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Homocysteine sulfinic acid (HCSA) is a homologue of the amino acid cysteine and a selective metabotropic glutamate receptor (mGluR) agonist. However, the metabolic role of HCSA is poorly understood. In this study, we showed that HCSA and glutamate stimulated glucose uptake in C2C12 mouse myoblast cells and increased AMP-activated protein kinase (AMPK) phosphorylation. RT-PCR and Western blot analysis revealed that C2C12 expresses mGluR5. HCSA transiently increased the intracellular calcium concentration. Although α-methyl-4-carboxyphenylglycine, a metabotropic glutamate receptor antagonist, blocked the action of HCSA in intracellular calcium response and AMPK phosphorylation, 6-cyano-7-nitroquinoxaline-2,3-dione, an AMPA antagonist, did not exhibit such effects. Knockdown of mGluR5 with siRNA blocked HCSA-induced AMPK phosphorylation. Pretreatment of cells with STO-609, a calmodulin-dependent protein kinase kinase (CaMKK) inhibitor, blocked HCSA-induced AMPK phosphorylation, and knockdown of CaMKK blocked HCSA-induced AMPK phosphorylation. In addition, HCSA activated p38 mitogen-activated protein kinase (MAPK). Expression of dominant-negative AMPK suppressed HCSA-mediated phosphorylation of p38 MAPK, and inhibition of AMPK and p38 MAPK blocked HCSA-induced glucose uptake. Phosphorylation of protein kinase C ζ (PKCζ) was also increased by HCSA. Pharmacologic inhibition or knockdown of p38 MAPK blocked HCSA-induced PKCζ phosphorylation, and knockdown of PKCζ suppressed the HCSA-induced increase of cell surface GLUT4. The stimulatory effect of HCSA on cell surface GLUT4 was impaired in FITC-conjugated PKCζ siRNA-transfected cells. Together, the above results suggest that HCSA may have a beneficial role in glucose metabolism in skeletal muscle cells via stimulation of AMPK.
Collapse
Affiliation(s)
- Ji Hae Kim
- Department of Anatomy, Korea University College of Medicine, Seoul 136-701, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Fallarino F, Volpi C, Fazio F, Notartomaso S, Vacca C, Busceti C, Bicciato S, Battaglia G, Bruno V, Puccetti P, Fioretti MC, Nicoletti F, Grohmann U, Di Marco R. Metabotropic glutamate receptor-4 modulates adaptive immunity and restrains neuroinflammation. Nat Med 2010; 16:897-902. [PMID: 20657581 DOI: 10.1038/nm.2183] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Accepted: 06/21/2010] [Indexed: 12/13/2022]
Abstract
High amounts of glutamate are found in the brains of people with multiple sclerosis, an inflammatory disease marked by progressive demyelination. Glutamate might affect neuroinflammation via effects on immune cells. Knockout mice lacking metabotropic glutamate receptor-4 (mGluR4) were markedly vulnerable to experimental autoimmune encephalomyelitis (EAE, a mouse model of multiple sclerosis) and developed responses dominated by interleukin-17-producing T helper (T(H)17) cells. In dendritic cells (DCs) from those mice, defective mGluR4 signaling-which would normally decrease intracellular cAMP formation-biased T(H) cell commitment to the T(H)17 phenotype. In wild-type mice, mGluR4 was constitutively expressed in all peripheral DCs, and this expression increased after cell activation. Treatment of wild-type mice with a selective mGluR4 enhancer increased EAE resistance via regulatory T (T(reg)) cells. The high amounts of glutamate in neuroinflammation might reflect a counterregulatory mechanism that is protective in nature and might be harnessed therapeutically for restricting immunopathology in multiple sclerosis.
Collapse
Affiliation(s)
- Francesca Fallarino
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Perugia, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Expression of NMDA receptor subunits in human peripheral blood lymphocytes in opioid addiction. Eur J Pharmacol 2010; 638:29-32. [DOI: 10.1016/j.ejphar.2010.04.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 03/15/2010] [Accepted: 04/13/2010] [Indexed: 11/21/2022]
|
26
|
Sedaghati M, Vousooghi N, Goodarzi A, Yaghmaei P, Mokri A, Zarrindast MR. Expression of NR3B but not NR2D subunit of NMDA receptor in human blood lymphocytes can serve as a suitable peripheral marker for opioid addiction studies. Eur J Pharmacol 2010; 633:50-4. [PMID: 20153313 DOI: 10.1016/j.ejphar.2010.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 12/17/2009] [Accepted: 02/02/2010] [Indexed: 11/18/2022]
Abstract
Glutamate is critically involved in opioid addiction. It has been suggested that neurotransmitter receptors expression in peripheral blood lymphocytes may reflect brain status. In the present study, using Real-time PCR, the mRNA expression of NR2D and NR3B subunits of NMDA glutamate receptor has been investigated in peripheral blood lymphocytes of four groups each comprising of 25 male individuals: opioid addicts, methadone maintained patients, long-term abstinent former opioid addicts, and non-addicted control subjects. We found that NR2D subunit mRNA expression was not changed in all three test groups in comparison to control subjects. However, the NR3B mRNA expression was significantly up-regulated by the factors 9.11 (P<0.001), 10.07 (P<0.001) and 4.08 (P<0.05) in abstinent, addicted and methadone maintained subjects, respectively relative to control group. As a conclusion, our data indicate that the transcriptional level of the NR2D subunit of NMDA receptor is not regulated by chronic opioid addiction. However, it seems that the over-expression of NR3B subunit of NMDA receptor is a long lasting result of opioid abuse. In addition, considerable decrease in the up-regulated state of the NR3B subunit by methadone may represent another benefit of methadone therapy for opioid addicts and may serve as a suitable marker to evaluate the successfulness of therapy.
Collapse
Affiliation(s)
- Mahmoud Sedaghati
- Science and Research Branch of Islamic Azad University, Tehran, Iran; Sina Cellular and Molecular Research Center, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
27
|
Pshenichkin S, Dolińska M, Klauzińska M, Luchenko V, Grajkowska E, Wroblewski JT. Dual neurotoxic and neuroprotective role of metabotropic glutamate receptor 1 in conditions of trophic deprivation - possible role as a dependence receptor. Neuropharmacology 2008; 55:500-8. [PMID: 18619982 PMCID: PMC2606294 DOI: 10.1016/j.neuropharm.2008.06.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 06/04/2008] [Accepted: 06/12/2008] [Indexed: 12/22/2022]
Abstract
Group I metabotropic glutamate receptors have been often implicated in various models of neuronal toxicity, however, the role played by the individual receptors and their putative mechanisms of action contributing to neurotoxicity or neuroprotection remain unclear. Here, using primary cultures of rat cerebellar granule cells and mouse cortical neurons, we show that conditions of trophic deprivation increased mGlu1 expression which correlated with the developing cell death. The inhibition of mGlu1 expression by specific siRNA attenuated toxicity, while adenovirus-mediated overexpression of mGlu1 resulted in increased cell death, indicating a causal relationship between the level of receptor expression and neuronal survival. In pharmacological experiments selective mGlu1 antagonists failed to protect from mGlu1-induced cell death, instead, neuronal survival was promoted by glutamate acting at mGlu1 receptors. Such properties are characteristics of a novel heterogeneous family of dependence receptors which control neuronal apoptosis. Our findings indicate that increased expression of mGlu1 in neurons creates a state of cellular dependence on the presence of its endogenous agonist glutamate. We propose a new role and a new mechanism for mGlu1 action. This receptor may play a crucial role in determining the fate of individual neurons during the development of the nervous system.
Collapse
Affiliation(s)
- Sergey Pshenichkin
- Department of Pharmacology, Georgetown University Medical Center, 3900 Reservoir Road NW, Washington, DC 20057, United States
| | | | | | | | | | | |
Collapse
|
28
|
Cizeron-Clairac G, Le Panse R, Frenkian-Cuvelier M, Meraouna A, Truffault F, Bismuth J, Mussot S, Kerlero de Rosbo N, Berrih-Aknin S. Thymus and Myasthenia Gravis: What can we learn from DNA microarrays? J Neuroimmunol 2008; 201-202:57-63. [DOI: 10.1016/j.jneuroim.2008.06.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2008] [Accepted: 06/12/2008] [Indexed: 11/16/2022]
|
29
|
Shin SS, Martino JJ, Chen S. Metabotropic glutamate receptors (mGlus) and cellular transformation. Neuropharmacology 2008; 55:396-402. [PMID: 18554669 PMCID: PMC2605077 DOI: 10.1016/j.neuropharm.2008.04.021] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Accepted: 04/24/2008] [Indexed: 12/20/2022]
Abstract
Although the glutamatergic system usually functions in the CNS, expression has been observed in non-neuronal tissues and a subset of cancers. Metabotropic glutamate receptors (mGlus) are highly "druggable" GPCRs and thus a priority for validation as therapeutic targets. We have previously reported that the aberrant expression of mGlu1 is sufficient to induce spontaneous melanoma development in vivo. We isolated and characterized several stable mGlu1-mouse melanocytic clones and demonstrated that these clones are transformed and tumorigenic. We hypothesize that expression of mGlus may not be uncommon in the pathogenesis of tumors other than melanoma, and that activity of an otherwise normal glutamate receptor in an ectopic cellular environment involves signaling pathways which dysregulate cell growth, ultimately leading to tumorigenesis. As most human cancers are of epithelial origin (carcinomas), in this review, the possibility that mGlu1 could function as a complete oncogene and transform epithelial cells is also discussed.
Collapse
Affiliation(s)
- Seung-Shick Shin
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, 164 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
30
|
Rousseaux CG. A Review of Glutamate Receptors II: Pathophysiology and Pathology. J Toxicol Pathol 2008. [DOI: 10.1293/tox.21.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Colin G. Rousseaux
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa
| |
Collapse
|
31
|
Taguchi K, Tamba M, Bannai S, Sato H. Induction of cystine/glutamate transporter in bacterial lipopolysaccharide induced endotoxemia in mice. JOURNAL OF INFLAMMATION-LONDON 2007; 4:20. [PMID: 17897437 PMCID: PMC2039726 DOI: 10.1186/1476-9255-4-20] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Accepted: 09/26/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND Cystine/glutamate transporter, system xc-, contributes to the maintenance of intracellular glutathione levels and the redox balance in the extracellular space. The main component of the transporter, xCT, is known to be strongly induced by various stimuli like oxidative stress in mammalian cultured cells. We examined the expression of xCT mRNA in vivo in the experimental endotoxemia. METHODS Northern blot analysis and in situ hybridization were used to investigate the expression of xCT mRNA in the tissues of the mice exposed to bacterial lipopolysaccharide (LPS). RESULTS Northern blot analysis revealed that xCT mRNA was constitutively expressed in the brain, thymus, and spleen, and that the expression of xCT mRNA was strongly up-regulated in thymus and spleen by the administration of a sublethal dose of LPS. In addition to brain, thymus, and spleen, xCT mRNA was detected also in the bronchiolar epithelium of the lung by the administration of the lethal dose of LPS. CONCLUSION xCT is induced in some specific tissues by the administration of LPS. The results suggest that cystine/glutamate transporter plays an important role under the inflammatory conditions.
Collapse
Affiliation(s)
- Kumiko Taguchi
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Michiko Tamba
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shiro Bannai
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hideyo Sato
- Department of Bioresource Engineering, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| |
Collapse
|
32
|
Changes in neurotransmitter levels and proinflammatory cytokine mRNA expressions in the mice olfactory bulb following nanoparticle exposure. Toxicol Appl Pharmacol 2007; 226:192-8. [PMID: 17950771 DOI: 10.1016/j.taap.2007.09.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 09/12/2007] [Accepted: 09/13/2007] [Indexed: 01/15/2023]
Abstract
Recently, there have been increasing reports that nano-sized component of particulate matter can reach the brain and may be associated with neurodegenerative diseases. Previously, our laboratory has studied the effect of intranasal instillation of nano-sized carbon black (CB) (14 nm and 95 nm) on brain cytokine and chemokine mRNA expressions and found that 14-nm CB increased IL-1 beta, TNF-alpha, CCL2 and CCL3 mRNA expressions in the olfactory bulb, not in the hippocampus of mice. To investigate the effect of a single administration of nanoparticles on neurotransmitters and proinflammatory cytokines in a mouse olfactory bulb, we performed in vivo microdialysis and real-time PCR methods. Ten-week-old male BALB/c mice were implanted with guide cannula in the right olfactory bulb and, 1 week later, were instilled vehicle or CB (14 nm, 250 microg) intranasally. Six hours after the nanoparticle instillation, the mice were intraperitoneally injected with normal saline or 50 mug of bacteria cell wall component lipoteichoic acid (LTA), which may potentiate CB-induced neurologic effect. Extracellular glutamate and glycine levels were significantly increased in the olfactory bulb of CB-instilled mice when compared with vehicle-instilled control mice. Moreover, we found that LTA further increased glutamate and glycine levels. However, no alteration of taurine and GABA levels was observed in the olfactory bulb of the same mice. We also detected immunological changes in the olfactory bulb 11 h after vehicle or CB instillation and found that IL-1 beta mRNA expression was significantly increased in CB- and LTA-treated mice when compared with control group. However, TNF-alpha mRNA expression was increased significantly in CB- and saline-treated mice when compared with control group. These findings suggest that nanoparticle CB may modulate the extracellular amino acid neurotransmitter levels and proinflammatory cytokine IL-1 beta mRNA expressions synergistically with LTA in the mice olfactory bulb.
Collapse
|
33
|
Miglio G, Dianzani C, Fallarini S, Fantozzi R, Lombardi G. Stimulation of N-methyl-d-aspartate receptors modulates Jurkat T cell growth and adhesion to fibronectin. Biochem Biophys Res Commun 2007; 361:404-9. [PMID: 17662248 DOI: 10.1016/j.bbrc.2007.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 07/04/2007] [Indexed: 12/31/2022]
Abstract
The aims of this study were to investigate the expression and the functional roles of N-methyl-d-aspartate (NMDA) receptors in leukemic Jurkat T cells. RT-PCR and immunofluorescence/confocal microscopy analysis showed that Jurkat T cells express the NR1 and NR2B subunits of the NMDA receptors. Exposure of Jurkat cells to either (5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,b]cyclohepten-5,10-imine [(+)-MK 801] or D-(-)-2-amino-5-phosphonopentanoic acid (D-AP5), two selective NMDA receptor antagonists, limited cell growth by inhibiting cell cycle progression and inducing apoptosis, whereas l-glutamate (1 microM) and NMDA (10 microM) significantly increased (137.2+/-22.0%; P<0.01) Jurkat T cell adhesion to fibronectin. In conclusion, our results demonstrate that Jurkat T cells express NMDA receptors functionally active in controlling cell growth and adhesion to fibronectin.
Collapse
Affiliation(s)
- Gianluca Miglio
- Department of Anatomy, Pharmacology and Forensic Medicine, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy
| | | | | | | | | |
Collapse
|
34
|
Nicoletti F, Battaglia G, Storto M, Ngomba RT, Iacovelli L, Arcella A, Gradini R, Sale P, Rampello L, De Vita T, Di Marco R, Melchiorri D, Bruno V. Metabotropic glutamate receptors: beyond the regulation of synaptic transmission. Psychoneuroendocrinology 2007; 32 Suppl 1:S40-5. [PMID: 17651904 DOI: 10.1016/j.psyneuen.2007.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 04/24/2007] [Indexed: 12/29/2022]
Abstract
Metabotropic glutamate (mGlu) receptors are G-protein coupled receptors activated by glutamate, the major excitatory neurotransmitter of the CNS. A growing body of evidence suggests that the function of mGlu receptors is not restricted to the regulation of synaptic transmission. mGlu receptors are expressed in a variety of peripheral cells, including inter alia hepatocytes, pancreatic cells, osteoblasts and immune cells. Within the immunological synapses, mGlu receptors expressed by T cells might contribute to the vast array of signals generated by the antigen-presenting cells. mGlu receptors are also found in embryonic and neural stem cells. This suggests their involvement in the pathophysiology of brain tumors, which likely originates from cancer stem cells similar to neural stem cells. Ligands of mGlu3 and mGlu4 receptors are potential candidates for the experimental treatment of malignant gliomas and medulloblastomas, respectively.
Collapse
Affiliation(s)
- Ferdinando Nicoletti
- Department of Human Physiology and Pharmacology, University of Rome La Sapienza, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Pavlovic V, Pavlovic D, Kocic G, Sokolovic D, Jevtovic-Stoimenov T, Cekic S, Velickovic D. Effect of monosodium glutamate on oxidative stress and apoptosis in rat thymus. Mol Cell Biochem 2007; 303:161-6. [PMID: 17458517 DOI: 10.1007/s11010-007-9469-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2007] [Accepted: 03/30/2007] [Indexed: 12/01/2022]
Abstract
It has been demonstrated that administration of high concentrations of monosodium glutamate (MSG), induce oxidative stress in different organs, but not in thymus. In the present study we examined the role of oxidative stress in MSG-induced thymocyte apoptosis. MSG was administrated intraperitoneally (4 mg/g of body weight) for six consecutive days. Animals were sacrificed at 1st, 7th, and 15th day after last MSG dose. MSG administration to animals significantly increased apoptotic rate of thymocytes (P < 0.01), together with significant increase of malondialdehyde (MDA) level (P < 0.001) and xanthine oxidase (XO) activity (P < 0.01), in time dependent manner. Catalase activity, during examination period, was significantly decreased (0 < 0.01). Obtained results showed that MSG treatment induced oxidative stress in thymus, which may have an important role in thymocyte apoptosis induced by MSG.
Collapse
Affiliation(s)
- Voja Pavlovic
- Institute of Physiology, Medical Faculty University of Nis, Bulevar dr Zorana Djindjica, Nis, Serbia.
| | | | | | | | | | | | | |
Collapse
|
36
|
Le Panse R, Cizeron-Clairac G, Bismuth J, Berrih-Aknin S. Microarrays reveal distinct gene signatures in the thymus of seropositive and seronegative myasthenia gravis patients and the role of CC chemokine ligand 21 in thymic hyperplasia. THE JOURNAL OF IMMUNOLOGY 2007; 177:7868-79. [PMID: 17114458 PMCID: PMC1892191 DOI: 10.4049/jimmunol.177.11.7868] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Myasthenia gravis (MG) is an autoimmune disease mainly caused by antiacetylcholine receptor autoantibodies (seropositive (SP) disease) or by Abs against unknown autoantigenic target(s) (seronegative (SN) disease). Thymectomy is usually beneficial although thymic hyperplasia with ectopic germinal centers is mainly observed in SP MG. To understand the role of thymus in the disease process, we compared the thymic transcriptome of non-MG adults to those of SP patients with a low or high degree of hyperplasia or SN patients. Surprisingly, an overexpression of MHC class II, Ig, and B cell marker genes is observed in SP but also SN MG patients. Moreover, we demonstrate an overexpression of CXCL13 in all MG thymuses leading probably to the generalized B cell infiltration. However, we find different chemotactic properties for MG subgroups and, especially, a specific overexpression of CCL21 in hyperplastic thymuses triggering most likely ectopic germinal center development. Besides, SN patients present a peculiar signature with an abnormal expression of genes involved in muscle development and synaptic transmission, but also genes implicated in host response, suggesting that viral infection might be related to SN MG. Altogether, these results underline differential pathogenic mechanisms in the thymus of SP and SN MG and propose new research areas.
Collapse
Affiliation(s)
- Rozen Le Panse
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8162, Institut Paris-Sud Cytokines, Université Paris XI, Hôpital Marie Lannelongue, 133 avenue de la résistance, 92350 Le Plessis-Robinson, France
| | | | | | | |
Collapse
|
37
|
Pacheco R, Oliva H, Martinez-Navío JM, Climent N, Ciruela F, Gatell JM, Gallart T, Mallol J, Lluis C, Franco R. Glutamate Released by Dendritic Cells as a Novel Modulator of T Cell Activation. THE JOURNAL OF IMMUNOLOGY 2006; 177:6695-704. [PMID: 17082582 DOI: 10.4049/jimmunol.177.10.6695] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adaptive immune responses begin after productive immunosynaptic contacts formation established in secondary lymphoid organs by dendritic cells (DC) presenting the Ag to T lymphocytes. Despite its resemblance to the neurosynapse, the participation of soluble small nonpeptidic mediators in the intercellular cross-talk taking place during T cell-DC interactions remains poorly studied. In this study, we show that human DC undergoing maturation and in contact with T cells release significant amounts of glutamate, which is the main excitatory neurotransmitter in mammalians. The release of glutamate is nonvesicular and mediated by the DC-expressed Xc- cystine/glutamate antiporter. DC-derived glutamate stimulating the constitutively expressed metabotropic glutamate receptor 5 impairs T cell activation. However, after productive Ag presentation, metabotropic glutamate receptor 1 is expressed in T cells to mediate enhanced T cell proliferation and secretion of Th1 and proinflammatory cytokines. These data suggest that, during T cell-DC interaction, glutamate is a novel and highly effective regulator in the initiation of T cell-mediated immune responses.
Collapse
Affiliation(s)
- Rodrigo Pacheco
- Department of Biochemistry and Molecular Biology, Faculty of Biology and Institut d'Investigació Biomèdica August Pi i Sunyer, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ferraguti F, Shigemoto R. Metabotropic glutamate receptors. Cell Tissue Res 2006; 326:483-504. [PMID: 16847639 DOI: 10.1007/s00441-006-0266-5] [Citation(s) in RCA: 402] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2006] [Accepted: 05/31/2006] [Indexed: 12/22/2022]
Abstract
Metabotropic glutamate receptors (mGlus) are a family of G-protein-coupled receptors activated by the neurotransmitter glutamate. Molecular cloning has revealed eight different subtypes (mGlu1-8) with distinct molecular and pharmacological properties. Multiplicity in this receptor family is further generated through alternative splicing. mGlus activate a multitude of signalling pathways important for modulating neuronal excitability, synaptic plasticity and feedback regulation of neurotransmitter release. In this review, we summarize anatomical findings (from our work and that of other laboratories) describing their distribution in the central nervous system. Recent evidence regarding the localization of these receptors in peripheral tissues will also be examined. The distinct regional, cellular and subcellular distribution of mGlus in the brain will be discussed in view of their relationship to neurotransmitter release sites and of possible functional implications.
Collapse
Affiliation(s)
- Francesco Ferraguti
- Department of Pharmacology, Innsbruck Medical University, Peter Mayr Strasse 1a, A-6020, Innsbruck, Austria
| | | |
Collapse
|
39
|
Chiocchetti A, Miglio G, Mesturini R, Varsaldi F, Mocellin M, Orilieri E, Dianzani C, Fantozzi R, Dianzani U, Lombardi G. Group I mGlu receptor stimulation inhibits activation-induced cell death of human T lymphocytes. Br J Pharmacol 2006; 148:760-8. [PMID: 16751798 PMCID: PMC1617076 DOI: 10.1038/sj.bjp.0706746] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Accepted: 03/10/2006] [Indexed: 01/23/2023] Open
Abstract
1. The effects of L-glutamate on activation-induced cell death (AICD) of human activated (1 microg ml(-1) phytohemagglutinin plus 2 U ml(-1) interleukin-2; 8 days) T lymphocytes were studied by measuring anti-CD3 monoclonal antibody (10 microg ml(-1); 18 h)-induced cell apoptosis (Annexin V and propidium iodide staining). 2. L-Glutamate (1 x 10(-8)-1 x 10(-4) M) significantly (P < or = 0.01) inhibited AICD in a concentration-dependent manner (EC50=6.3 x 10(-8) M; maximum inhibition 54.8+/-6.3% at 1 x 10(-6) M). 3. The L-glutamate inhibitory effect was pharmacologically characterized as mediated by group I mGlu receptors, since mGlu receptor agonists reproduced this effect. The EC50 values were: 3.2 x 10(-7) M for (1S,3R)-ACPD; 4.5 x 10(-8) M for quisqualate; 1.0 x 10(-6) M for (S)-3,5-DHPG; 2.0 x 10(-5) M for CHPG. 4. Group I mGlu receptor antagonists inhibited the effects of quisqualate 1.0 x 10(-6) M. The IC50 values calculated were: 8.7 x 10(-5), 4.3 x 10(-6) and 6.3 x 10(-7) M for AIDA, LY 367385 and MPEP, respectively. 5. L-Glutamate (1 x 10(-6) M; 18 h) significantly (P < or = 0.05) inhibited FasL expression (40.8+/-11.3%) (cytofluorimetric analysis), whereas it did not affect Fas signalling. 6. Expression of both mGlu1 and mGlu5 receptor mRNA by T lymphocytes and T-cell lines, as demonstrated by reverse transcriptase-PCR analysis, suggests that L-glutamate-mediated inhibition of AICD was exerted on T cells. 7. These data depict a novel role for L-glutamate in the regulation of the immune response through group I mGlu receptor-mediated mechanisms.
Collapse
Affiliation(s)
- Annalisa Chiocchetti
- Interdisciplinary Research Center of Autoimmune Diseases, Department of Medical Sciences, Eastern Piedmont University, Via Solaroli, 17, 28100 Novara, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Storto M, Capobianco L, Battaglia G, Molinaro G, Gradini R, Riozzi B, Di Mambro A, Mitchell KJ, Bruno V, Vairetti MP, Rutter GA, Nicoletti F. Insulin secretion is controlled by mGlu5 metabotropic glutamate receptors. Mol Pharmacol 2006; 69:1234-41. [PMID: 16424079 DOI: 10.1124/mol.105.018390] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recent evidence suggests that metabotropic glutamate (mGlu) receptors are involved in the regulation of hormone secretion in the endocrine pancreas. We report here that endogenous activation of mGlu5 receptors is required for an optimal insulin response to glucose both in clonal beta-cells and in mice. In clonal beta-cells, mGlu5 receptors were expressed at the cell surface and were also found in purified insulin-containing granules. These cells did not respond to a battery of mGlu5 receptor agonists that act extracellularly, but instead responded to a cell-permeant analog of glutamate with an increase in [Ca2+]i and insulin secretion. Both effects were largely attenuated by the mGlu5 receptor antagonist, 2-methyl-6-(phenylethynyl)-pyridine (MPEP). MPEP and its structural analog, (E)-2-methyl-6-styryl-pyridine (SIB-1893), reduced the increase in [Ca2+]i and insulin secretion induced by glucose in clonal beta-cells, whereas a mGlu1 receptor antagonist was inactive. mGlu5 knockout mice showed a defective insulin response at all times after a glucose pulse (1.5 g/kg, i.p.), whereas wild-type mice treated with MPEP (10 mg/kg, i.p.) showed a selective impairment in the late phase of insulin secretion in response to glucose challenge. Mice injected with MPEP or lacking mGlu5 receptors also showed a blunted glucagon response to an insulin challenge. We conclude that insulin secretion is under the control of mGlu5 receptors both in clonal beta-cells and in vivo. Drugs that modulate the function of mGlu5 receptors might affect glucose homeostasis by altering the secretion of pancreatic hormones.
Collapse
|
41
|
Miglio G, Varsaldi F, Lombardi G. Human T lymphocytes express N-methyl-d-aspartate receptors functionally active in controlling T cell activation. Biochem Biophys Res Commun 2005; 338:1875-83. [PMID: 16289038 DOI: 10.1016/j.bbrc.2005.10.164] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2005] [Accepted: 10/25/2005] [Indexed: 10/25/2022]
Abstract
The aim of this study was to investigate the expression and the functional role of N-methyl-D-aspartate (NMDA) receptors in human T cells. RT-PCR analysis showed that human resting peripheral blood lymphocytes (PBL) and Jurkat T cells express genes encoding for both NR1 and NR2B subunits: phytohemagglutinin (PHA)-activated PBL also expresses both these genes and the NR2A and NR2D genes. Cytofluorimetric analysis showed that NR1 expression increases as a consequence of PHA (10 microg/ml) treatment. D-(-)-2-Amino-5-phosphonopentanoic acid (D-AP5), and (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine [(+)-MK 801], competitive and non-competitive NMDA receptor antagonists, respectively, inhibited PHA-induced T cell proliferation, whereas they did not affect IL-2 (10 U/ml)-induced proliferation of PHA blasts. These effects were due to the prevention of T cell activation (inhibition of cell aggregate formation and CD25 expression), but not to cell cycle arrest or death. These results demonstrate that human T lymphocytes express NMDA receptors, which are functionally active in controlling cell activation.
Collapse
Affiliation(s)
- Gianluca Miglio
- DiSCAFF Department, Eastern Piedmont University, Via Bovio 6, 28100 Novara, Italy
| | | | | |
Collapse
|
42
|
Boldyrev AA, Carpenter DO, Johnson P. Emerging evidence for a similar role of glutamate receptors in the nervous and immune systems. J Neurochem 2005; 95:913-8. [PMID: 16271044 DOI: 10.1111/j.1471-4159.2005.03456.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The role of glutamate receptors in synaptic transmission and excitotoxicity in the nervous system is well established. Recent evidence has emerged that glutamatergic mechanisms also exist in a wide variety of non-neuronal cells. In the case of thymocytes and lymphocytes, several types of glutamate receptor are expressed which can induce functional changes. This review focuses on the cellular function of NMDA-activated ionotropic and groups I and III metabotropic glutamate receptors in lymphocytes. Levels of exogenous and endogenous circulatory agonists and antagonists for lymphocyte glutamate receptors, notably homocysteine metabolites, are markedly increased in certain disease states and may be involved in disorders of the immune system. In addition to glutamate and aspartate, these compounds are active at glutamate receptors and increase the excitotoxic effects of glutamate in both neurons and lymphocytes. Increased levels of compounds acting at glutamate receptors may be risk factors for organ damage, for example in both heart and kidney disease. We conclude that glutamate is involved in signaling in immunocompetent cells and that the expression of both ionotropic and metabotropic glutamate receptors may have regulatory functions in immunocompetent cells, as well as in the nervous system. In addition, glutamate may serve as a signaling agent between the immune and nervous systems.
Collapse
Affiliation(s)
- Alexander A Boldyrev
- Center for Molecular Medicine and International Biotechnological Center (Department of Biochemistry), M. V. Lomonosov Moscow State University, Moscow, Russia.
| | | | | |
Collapse
|
43
|
Miglio G, Varsaldi F, Dianzani C, Fantozzi R, Lombardi G. Stimulation of group I metabotropic glutamate receptors evokes calcium signals and c-jun and c-fos gene expression in human T cells. Biochem Pharmacol 2005; 70:189-99. [PMID: 15935992 DOI: 10.1016/j.bcp.2005.04.038] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Accepted: 04/13/2005] [Indexed: 11/28/2022]
Abstract
To study if the activation of group I mGlu receptors in human T cells modifies intracellular Ca2+ concentration ([Ca2+](i)) and cell function, we measured [Ca2+](i) on cell suspensions (spectrofluorimetric method) or single cell (digital Ca2+ imaging system) using fura-2 as indicator. Early-inducible gene (c-jun and c-fos) expression was studied by reverse transcriptase-polymerase chain reaction assay as representative of Ca(2+)-sensitive gene expression. (1S,3R)-ACPD (100 microM), the selective mGlu receptor agonist, evoked a significant increase (34.1+/-4.9%) of [Ca2+](i), pharmacologically characterized as mediated by group I mGlu receptors, since both (S)-3,5-DHPG (100 microM), a selective group I mGlu receptor agonist and CHPG (1mM), the specific mGlu5 receptor agonist, reproduced the effects, that were abolished by AIDA (1mM), a selective group I mGlu receptor antagonist. (S)-3,5-DHPG-induced a rapid [Ca2+](i) rise (initial phase) followed by a slow decrease (second phase) to the baseline. Both extracellular Ca2+ and Ca2+ released from intracellular stores contribute to the [Ca2+](i) increase which depend on PLC activation. In a Ca(2+)-free buffer, the second phase rapidly return to the baseline; LaCl3 (1-10 microM), an inhibitor of extracellular Ca2+ influx, significantly reduced the second phase only; thapsigargin (1microM), by discharging intracellular Ca2+ stores, U 73122 (10 microM) and D609 (300 microM), by inhibiting PLC activity, prevented both phases. In our system, PTX pre-treatment increased (S)-3,5-DHPG effects, demonstrating that PXT-sensitive G(i/o) proteins are involved. Finally, specific stimulation of these receptors in Jurkat cells upregulates c-jun and c-fos gene expression, thus activating multiple downstream signalling regulating important T cell functions.
Collapse
Affiliation(s)
- Gianluca Miglio
- DISCAFF, "Amedeo Avogadro" University of Eastern Piedmont, Via Bovio 6, 28100 Novara, Italy
| | | | | | | | | |
Collapse
|
44
|
Shaked I, Tchoresh D, Gersner R, Meiri G, Mordechai S, Xiao X, Hart RP, Schwartz M. Protective autoimmunity: interferon-gamma enables microglia to remove glutamate without evoking inflammatory mediators. J Neurochem 2005; 92:997-1009. [PMID: 15715651 DOI: 10.1111/j.1471-4159.2004.02954.x] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glutamate in excessive amounts is a major contributor to neuronal degeneration, and its removal is attributed mainly to astrocytes. Traumatic injury to the central nervous system (CNS) is often accompanied by disappearance of astrocytes from the lesion site and failure of the remaining cells to withstand the ensuing toxicity. Microglia that repopulate the lesion site are the usual suspects for causing redox imbalance and inflammation and thus further exacerbating the neurotoxicity. However, our group recently demonstrated that early post-injury activation of microglia as antigen-presenting cells correlates with an ability to withstand injurious conditions. Moreover, we found that T cells reactive to CNS-specific self-antigens protected neurons against glutamate toxicity. Here, we show that antigen-specific autoimmune T cells, by tailoring the microglial phenotype, can increase the ability of microglia-enriched cultures to remove glutamate. This T-cell-mediated effect could not be achieved by the potent microglia-activating agent lipopolysaccharide (LPS), but was dose-dependently reproduced by the Th1 cytokine interferon (IFN)-gamma and significantly reduced by neutralizing anti-IFN-gamma antibodies. Under the same conditions, IFN-gamma had no effect on cultured astrocytes. Up-regulation of glutamate uptake induced by IFN-gamma activation was not accompanied by the acute inflammatory response seen in LPS-activated cultures. These findings suggest that T cells or their cytokines can cause microglia to adopt a phenotype that facilitates rather than impairs glutamate clearance, possibly contributing to restoration of homeostasis.
Collapse
Affiliation(s)
- I Shaked
- Department of Neurobiology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Poulopoulou C, Markakis I, Davaki P, Nikolaou C, Poulopoulos A, Raptis E, Vassilopoulos D. Modulation of Voltage-Gated Potassium Channels in Human T Lymphocytes by Extracellular Glutamate. Mol Pharmacol 2005. [DOI: 10.1124/mol.67.3.856] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
46
|
Poulopoulou C, Davaki P, Koliaraki V, Kolovou D, Markakis I, Vassilopoulos D. Reduced expression of metabotropic glutamate receptor 2mRNA in T cells of ALS patients. Ann Neurol 2005; 58:946-9. [PMID: 16240362 DOI: 10.1002/ana.20675] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Metabotropic glutamate receptors alter the vulnerability of neurons to excitotoxic damage and are reported to display abnormal expression in the central nervous system of ALS patients. Using reverse transcriptase polymerase chain reaction, we investigated the mRNA expression of specific metabotropic glutamate receptor subtypes in T lymphocytes of 20 patients with sporadic ALS, compared with healthy age-matched control subjects and patients with other neurological disorders. The levels of metabotropic glutamate receptor 2 mRNA were markedly reduced, whereas the expression of other subtypes (1b, 3, 8) was similar to control levels. Our findings may provide a reliable peripheral marker of the glutamatergic dysfunction that characterizes ALS.
Collapse
Affiliation(s)
- Cornelia Poulopoulou
- Department of Neurology, Laboratory of Experimental Neurophysiology, Medical School, University of Athens, Eginition Hospital, 72-74 Vas. Sofias Avenue, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
47
|
Pacheco R, Ciruela F, Casadó V, Mallol J, Gallart T, Lluis C, Franco R. Group I metabotropic glutamate receptors mediate a dual role of glutamate in T cell activation. J Biol Chem 2004; 279:33352-8. [PMID: 15184389 DOI: 10.1074/jbc.m401761200] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Metabotropic glutamate receptors (mGluR) are present in cells of the nervous system, where they are activated by one of the main neurotransmitters, glutamate. They are also expressed in cells outside the nervous system. We identified and characterized two receptors belonging to group I mGluR, mGlu1R and mGlu5R, in human cell lines of lymphoid origin and in resting and activated lymphocytes from human peripheral blood. Both are highly expressed in the human Jurkat T cell line, whereas mGlu5R is expressed only in the human B cell line SKW6.4. In blood lymphocytes, mGlu5R is expressed constitutively, whereas mGlu1R is expressed only upon activation via the T cell receptor-CD3 complex. Group I receptors in the central nervous system are coupled to phospholipase C, whereas in blood lymphocytes, activation of mGlu5R does not trigger this signaling pathway, but instead activates adenylate cyclase. On the other hand, mGlu5R does not mediate ERK1/2 activation, whereas mGlu1R, which is coupled neither to phospholipase C nor to calcium channels and whose activation does not increase cAMP, activates the mitogen-activated protein kinase cascade. The differential expression of mGluR in resting and activated lymphocytes and the different signaling pathways that are triggered when mGlu1Rs or mGlu5Rs are activated point to a key role of glutamate in the regulation of T cell physiological function. The study of the signaling pathways (cAMP production and ERK1/2 phosphorylation) and the proliferative response obtained in the presence of glutamate analogs suggests that mGlu1R and mGlu5R have distinct functions. mGlu5R mediates the reported inhibition of cell proliferation evoked by glutamate, which is reverted by the activation of inducible mGlu1R. This is a novel non-inhibitory action mechanism for glutamate in lymphocyte activation. mGlu1R and mGlu5R thus mediate opposite glutamate effects in human lymphocytes.
Collapse
Affiliation(s)
- Rodrigo Pacheco
- Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
48
|
Lombardi G, Miglio G, Dianzani C, Mesturini R, Varsaldi F, Chiocchetti A, Dianzani U, Fantozzi R. Glutamate modulation of human lymphocyte growth: in vitro studies. Biochem Biophys Res Commun 2004; 318:496-502. [PMID: 15120628 DOI: 10.1016/j.bbrc.2004.04.053] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2004] [Indexed: 11/23/2022]
Abstract
Peripheral blood mononuclear cell (PBMC) proliferation induced by phytohemagglutinin, or by anti-CD3 alone or plus anti-CD28 monoclonal antibodies (mAb) was inhibited by glutamate (Glu) in a concentration-dependent manner. This inhibition was not reproduced by selective ionotropic Glu receptor agonists, whereas it was potentiated by l-buthionine-(S,R)-sulfoximine, which depletes glutathione (GSH) stores, and counteracted by 2-mercaptoethanol, a preserver of cell thiols. The inhibitory effects of Glu were related to depletion of intracellular GSH stores, since it decreased GSH levels in a concentration-dependent manner. Furthermore, Glu modulated cytokine secretion by anti-CD3 mAb activated PBMC: it increased IFN-gamma (+44.3+/-8.2%) and IL-10 (+31.6+/-9.7%) secretion, whereas that of IL-2, IL-4, IL-5, and TNF-alpha was not affected. These data suggest that high levels of Glu, which can be reached in damaged tissues, modulate lymphocyte responses to activating stimuli by favouring polarization of the T helper effector response.
Collapse
Affiliation(s)
- Grazia Lombardi
- Interdisciplinary Research Center of Autoimmune Diseases, DISCAFF Department, Eastern Piedmont University, Via Bovio 6, 28100 Novara, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Hinoi E, Takarada T, Ueshima T, Tsuchihashi Y, Yoneda Y. Glutamate signaling in peripheral tissues. ACTA ACUST UNITED AC 2004; 271:1-13. [PMID: 14686914 DOI: 10.1046/j.1432-1033.2003.03907.x] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The hypothesis that l-glutamate (Glu) is an excitatory amino acid neurotransmitter in the mammalian central nervous system is now gaining more support after the successful cloning of a number of genes coding for the signaling machinery required for this neurocrine at synapses in the brain. These include Glu receptors (signal detection), Glu transporters (signal termination) and vesicular Glu transporters (signal output through exocytotic release). Relatively little attention has been paid to the functional expression of these molecules required for Glu signaling in peripheral neuronal and non-neuronal tissues; however, recent molecular biological analyses show a novel function for Glu as an extracellular signal mediator in the autocrine and/or paracrine system. Emerging evidence suggests that Glu could play a dual role in mechanisms underlying the maintenance of cellular homeostasis - as an excitatory neurotransmitter in the central neurocrine system and an extracellular signal mediator in peripheral autocrine and/or paracrine tissues. In this review, the possible Glu signaling methods are outlined in specific peripheral tissues including bone, testis, pancreas, and the adrenal, pituitary and pineal glands.
Collapse
Affiliation(s)
- Eiichi Hinoi
- Laboratory of Molecular Pharmacology, Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Ishikawa, Japan
| | | | | | | | | |
Collapse
|
50
|
Arce C, Del Campo AB, Figueroa S, López E, Aránguez I, Oset-Gasque MJ, González MP. Expression and functional properties of group I metabotropic glutamate receptors in bovine chromaffin cells. J Neurosci Res 2003; 75:182-193. [PMID: 14705139 DOI: 10.1002/jnr.10824] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
We demonstrate the presence and functional properties of Group I metabotropic glutamate receptors (mGluRs) expressed in chromaffin cells. Immunocytochemical techniques revealed that two mGluR subtypes (mGluR1alpha and mGluR5) are expressed in chromaffin cells, located in both the cytoplasmic membrane and the cytosol surrounding the nucleus. These mGluRs are functionally active on catecholamine (CA) secretion in chromaffin cells because both (1S, 3R)-1-aminocyclopentane-1,3-dicarboxylic acid (t-ACPD) and the specific agonist of Group I mGluRs, (S)-3,5-dihydroxyphenylglycine (DHPG), were able to stimulate the release of CAs (adrenaline and noradrenaline) in a dose-response manner. These effects were specifically reversed by L-(+)-2-amino-3-phosphonopropionic acid (L-AP3), a selective antagonist of the Group I metabotropic glutamate receptors. t-ACPD induced an increase in CA secretion in both the presence and absence of extracellular calcium, the former effect being accompanied by cell membrane depolarization. Noradrenaline (NA) release was higher in the presence of extracellular calcium than in its absence, whereas adrenaline release was of the same order under both conditions. These results indicate that different subtypes of Group I mGluRs are present in noradrenergic and adrenergic cells. Fluorescence imaging techniques in single cells showed different t-ACPD-induced increases in intracellular calcium in different chromaffin cells: in chromaffin cells, 67% expressed functional metabotropic glutamate receptors and with nicotinic receptors, whereas the remaining 33% expressed only nicotinic receptors. In the absence of external calcium, only about 25% of cells responded to t-ACPD-increased intracellular calcium by increasing inositol 1,4,5-trisphosphate (IP(3)) concentration and subsequent calcium mobilization from intracellular stores, whereas the remaining 75% increased intracellular calcium by promoting Ca(2+) influx from the extracellular medium through L- and N- but not P/Q voltage-dependent calcium channels.
Collapse
Affiliation(s)
- C Arce
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - A B Del Campo
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - S Figueroa
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - E López
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - I Aránguez
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - M J Oset-Gasque
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| | - M P González
- Instituto de Bioquímica (Centro Mixto CSIC-UCM), Facultad de Farmacia, Madrid, Spain
| |
Collapse
|