101
|
Słoniecka M, Le Roux S, Boman P, Byström B, Zhou Q, Danielson P. Expression Profiles of Neuropeptides, Neurotransmitters, and Their Receptors in Human Keratocytes In Vitro and In Situ. PLoS One 2015. [PMID: 26214847 PMCID: PMC4516240 DOI: 10.1371/journal.pone.0134157] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Keratocytes, the quiescent cells of the corneal stroma, play a crucial role in corneal wound healing. Neuropeptides and neurotransmitters are usually associated with neuronal signaling, but have recently been shown to be produced also by non-neuronal cells and to be involved in many cellular processes. The aim of this study was to assess the endogenous intracellular and secreted levels of the neuropeptides substance P (SP) and neurokinin A (NKA), and of the neurotransmitters acetylcholine (ACh), catecholamines (adrenaline, noradrenaline and dopamine), and glutamate, as well as the expression profiles of their receptors, in human primary keratocytes in vitro and in keratocytes of human corneal tissue sections in situ. Cultured keratocytes expressed genes encoding for SP and NKA, and for catecholamine and glutamate synthesizing enzymes, as well as genes for neuropeptide, adrenergic and ACh (muscarinic) receptors. Keratocytes in culture produced SP, NKA, catecholamines, ACh, and glutamate, and expressed neurokinin-1 and -2 receptors (NK-1R and NK-2R), dopamine receptor D2, muscarinic ACh receptors, and NDMAR1 glutamate receptor. Human corneal sections expressed SP, NKA, NK-1R, NK-2R, receptor D2, choline acetyl transferase (ChAT), M3, M4 and M5 muscarinic ACh receptors, glutamate, and NMDAR1, but not catecholamine synthesizing enzyme or the α1 and β2 adrenoreceptors, nor M1 receptor. In addition, expression profiles assumed significant differences between keratocytes from the peripheral cornea as compared to those from the central cornea, as well as differences between keratocytes cultured under various serum concentrations. In conclusion, human keratocytes express an array of neuropeptides and neurotransmitters. The cells furthermore express receptors for neuropeptides/neurotransmitters, which suggests that they are susceptible to stimulation by these substances in the cornea, whether of neuronal or non-neuronal origin. As it has been shown that neuropeptides/neurotransmitters are involved in cell proliferation, migration, and angiogenesis, it is possible that they play a role in corneal wound healing.
Collapse
Affiliation(s)
- Marta Słoniecka
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- * E-mail:
| | - Sandrine Le Roux
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Peter Boman
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Berit Byström
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Qingjun Zhou
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Qingdao, China
| | - Patrik Danielson
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
102
|
Dean BJF, Snelling SJB, Dakin SG, Murphy RJ, Javaid MK, Carr AJ. Differences in glutamate receptors and inflammatory cell numbers are associated with the resolution of pain in human rotator cuff tendinopathy. Arthritis Res Ther 2015; 17:176. [PMID: 26160609 PMCID: PMC4498529 DOI: 10.1186/s13075-015-0691-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/18/2015] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION The relationship between peripheral tissue characteristics and pain symptoms in soft tissue inflammation is poorly understood. The primary aim of this study was to determine immunohistochemical differences in tissue obtained from patients with persistent pain and patients who had become pain-free after surgical treatment for rotator cuff tendinopathy. The secondary aim was to investigate whether there would be differences in glutaminergic and inflammatory gene expression between disease-derived and healthy control cells in vitro. METHODS Supraspinatus tendon biopsies were obtained from nine patients with tendon pain before shoulder surgery and from nine further patients whose pain had resolved completely following shoulder surgery. Histological markers relating to the basic tendon characteristics, inflammation and glutaminergic signalling were quantified by immunohistochemical analysis. Gene expression of glutaminergic and inflammatory markers was determined in tenocyte explants derived from painful rotator cuff tendon tears in a separate cohort of patients and compared to that of explants from healthy control tendons. Dual labelling was performed to identify cell types expressing nociceptive neuromodulators. RESULTS Tendon samples from patients with persistent pain demonstrated increased levels of metabotropic glutamate receptor 2 (mGluR2), kainate receptor 1 (KA1), protein gene product 9.5 (PGP9.5), CD206 (macrophage marker) and CD45 (pan-leucocyte marker) versus pain-free controls (p <0.05). NMDAR1 co-localised with CD206-positive cells, whereas PGP9.5 and glutamate were predominantly expressed by resident tendon cells. These results were validated by in vitro increases in the expression of mGluR2, N-methyl-D-aspartate receptor (NMDAR1), KA1, CD45, CD206 and tumour necrosis factor alpha (TNF-α) genes (p <0.05) in disease-derived versus control cells. CONCLUSIONS We conclude that differences in glutamate receptors and inflammatory cell numbers are associated with the resolution of shoulder pain in rotator cuff tendinopathy, and that disease-derived cells exhibit a distinctly different neuro-inflammatory gene expression profile to healthy control cells.
Collapse
Affiliation(s)
- Benjamin John Floyd Dean
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Sarah J B Snelling
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Richard J Murphy
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Muhammad Kassim Javaid
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| | - Andrew Jonathan Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, OX3 7LD, UK.
| |
Collapse
|
103
|
Gregory KJ, Conn PJ. Molecular Insights into Metabotropic Glutamate Receptor Allosteric Modulation. Mol Pharmacol 2015; 88:188-202. [PMID: 25808929 PMCID: PMC4468636 DOI: 10.1124/mol.114.097220] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/24/2015] [Indexed: 12/21/2022] Open
Abstract
The metabotropic glutamate (mGlu) receptors are a group of eight family C G protein-coupled receptors that are expressed throughout the central nervous system (CNS) and periphery. Within the CNS the different subtypes are found in neurons, both pre- and/or postsynaptically, where they mediate modulatory roles and in glial cells. The mGlu receptor family provides attractive targets for numerous psychiatric and neurologic disorders, with the majority of discovery programs focused on targeting allosteric sites, with allosteric ligands now available for all mGlu receptor subtypes. However, the development of allosteric ligands remains challenging. Biased modulation, probe dependence, and molecular switches all contribute to the complex molecular pharmacology exhibited by mGlu receptor allosteric ligands. In recent years we have made significant progress in our understanding of this molecular complexity coupled with an increased understanding of the structural basis of mGlu allosteric modulation.
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Vanderbilt Center for Neuroscience Drug Discovery & Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (P.J.C)
| | - P Jeffrey Conn
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia (K.J.G.); and Vanderbilt Center for Neuroscience Drug Discovery & Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee (P.J.C)
| |
Collapse
|
104
|
Discovery and biological evaluation of tetrahydrothieno[2,3-c]pyridine derivatives as selective metabotropic glutamate receptor 1 antagonists for the potential treatment of neuropathic pain. Eur J Med Chem 2015; 97:245-58. [DOI: 10.1016/j.ejmech.2015.04.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/30/2015] [Accepted: 04/28/2015] [Indexed: 12/31/2022]
|
105
|
Gammon JM, Tostanoski LH, Adapa AR, Chiu YC, Jewell CM. Controlled delivery of a metabolic modulator promotes regulatory T cells and restrains autoimmunity. J Control Release 2015; 210:169-78. [PMID: 26002150 DOI: 10.1016/j.jconrel.2015.05.277] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 05/16/2015] [Accepted: 05/18/2015] [Indexed: 12/13/2022]
Abstract
Autoimmune disorders occur when the immune system abnormally recognizes and attacks self-molecules. Dendritic cells (DCs) play a powerful role in initiating adaptive immune response, and are therefore a recent target for autoimmune therapies. N-Phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC), a small molecule glutamate receptor enhancer, alters how DCs metabolize glutamate, skewing cytokine secretion to bias T cell function. These effects provide protection in mouse models of multiple sclerosis (MS) by polarizing T cells away from inflammatory TH17 cells and toward regulatory T cells (TREG) when mice receive daily systemic injections of PHCCC. However, frequent, continued treatment is required to generate and maintain therapeutic benefits. Thus, the use of PHCCC is limited by poor solubility, the need for frequent dosing, and cell toxicity. We hypothesized that controlled release of PHCCC from degradable nanoparticles (NPs) might address these challenges by altering DC function to maintain efficacy with reduced treatment frequency and toxicity. This idea could serve as a new strategy for harnessing biomaterials to polarize immune function through controlled delivery of metabolic modulators. PHCCC was readily encapsulated in nanoparticles, with controlled release of 89% of drug into media over three days. Culture of primary DCs or DC and T cell co-cultures with PHCCC NPs reduced DC activation and secretion of pro-inflammatory cytokines, while shifting T cells away from TH17 and toward TREG phenotypes. Importantly, PHCCC delivered to cells in NPs was 36-fold less toxic compared with soluble PHCCC. Treatment of mice with PHCCC NPs every three days delayed disease onset and decreased disease severity compared with mice treated with soluble drug at the same dose and frequency. These results highlight the potential to promote tolerance through controlled delivery of metabolic modulators that alter DC signaling to polarize T cells, and suggest future gains that could be realized by engineering materials that provide longer term release.
Collapse
Affiliation(s)
- Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Arjun R Adapa
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Yu-Chieh Chiu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, United States; Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, United States; Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, United States.
| |
Collapse
|
106
|
Anaparti V, Ilarraza R, Orihara K, Stelmack GL, Ojo OO, Mahood TH, Unruh H, Halayko AJ, Moqbel R. NMDA receptors mediate contractile responses in human airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1253-64. [PMID: 25888577 DOI: 10.1152/ajplung.00402.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/15/2015] [Indexed: 01/12/2023] Open
Abstract
Human airway smooth muscle (HASM) exhibits enhanced contractility in asthma. Inflammation is associated with airway hypercontractility, but factors that underpin these features are not fully elucidated. Glutamate toxicity associated with increased plasma glutamate concentrations was observed in airway inflammation, suggesting that multisubunit glutamate receptors, N-methyl-d-aspartate receptors (NMDA-R) contribute to airway hyperreactivity. We tested the hypothesis that HASM expresses NMDA-R subunits that can form functional receptors to mediate contractile responses to specific extracellular ligands. In cultured HASM cells, we measured NMDA-R subunit mRNA and protein abundance by quantitative PCR, immunoblotting, flow cytometry, and epifluorescence immunocytochemistry. We measured mRNA for a number of NMDA-R subunits, including the obligatory NR1 subunit, which we confirmed to be present as a protein. In vitro and ex vivo functional NMDA-R activation in HASM cells was measured using intracellular calcium flux (fura-2 AM), collagen gel contraction assays, and murine thin-cut lung slices (TCLS). NMDA, a pharmacological glutamate analog, induced cytosolic calcium mobilization in cultured HASM cells. We detected three different temporal patterns of calcium response, suggesting the presence of heterogeneous myocyte subpopulations. NMDA-R activation also induced airway contraction in murine TCLS and soft collagen gels seeded with HASM cells. Responses in cells, lung slices, and collagen gels were mediated by NMDA-R, as they could be blocked by (2R)-amino-5-phosphonopentanoate, a specific NMDA-R inhibitor. In summary, we reveal the presence of NMDA-R in HASM that mediate contractile responses via glutamatergic mechanisms. These findings suggest that accumulation of glutamate-like ligands for NMDA-R associated with airway inflammation contributes directly to airway hyperreactivity.
Collapse
Affiliation(s)
- Vidyanand Anaparti
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Ramses Ilarraza
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Kanami Orihara
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gerald L Stelmack
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Oluwaseun O Ojo
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Thomas H Mahood
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Helmut Unruh
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Section of Thoracic Surgery, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada;
| | - Redwan Moqbel
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada; Biology of Breathing Group, Child Health Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
107
|
Moloney RD, Golubeva AV, O'Connor RM, Kalinichev M, Dinan TG, Cryan JF. Negative allosteric modulation of the mGlu7 receptor reduces visceral hypersensitivity in a stress-sensitive rat strain. Neurobiol Stress 2015; 2:28-33. [PMID: 26844237 PMCID: PMC4721404 DOI: 10.1016/j.ynstr.2015.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 03/15/2015] [Accepted: 04/03/2015] [Indexed: 11/19/2022] Open
Abstract
Glutamate, the main excitatory neurotransmitter in the central nervous system, exerts its effect through ionotropic and metabotropic receptors. Of these, group III mGlu receptors (mGlu 4, 6, 7, 8) are among the least studied due to a lack of pharmacological tools. mGlu7 receptors, the most highly conserved isoform, are abundantly distributed in the brain, especially in regions, such as the amygdala, known to be crucial for the emotional processing of painful stimuli. Visceral hypersensitivity is a poorly understood phenomenon manifesting as an increased sensitivity to visceral stimuli. Glutamate has long been associated with somatic pain processing leading us to postulate that crossover may exist between these two modalities. Moreover, stress has been shown to exacerbate visceral pain. ADX71743 is a novel, centrally penetrant, negative allosteric modulator of mGlu7 receptors. Thus, we used this tool to explore the possible involvement of this receptor in the mediation of visceral pain in a stress-sensitive model of visceral hypersensitivity, namely the Wistar Kyoto (WKY) rat. ADX71743 reduced visceral hypersensitivity in the WKY rat as exhibited by increased visceral sensitivity threshold with concomitant reductions in total number of pain behaviours. Moreover, AD71743 increased total distance and distance travelled in the inner zone of the open field. These findings show, for what is to our knowledge, the first time, that mGlu7 receptor signalling plays a role in visceral pain processing. Thus, negative modulation of the mGlu7 receptor may be a plausible target for the amelioration of stress-induced visceral pain where there is a large unmet medical need.
Collapse
Affiliation(s)
- Rachel D. Moloney
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Psychiatry, University College Cork, Ireland
| | - Anna V. Golubeva
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
| | | | | | - Timothy G. Dinan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Psychiatry, University College Cork, Ireland
| | - John F. Cryan
- Laboratory of Neurogastroenterology, Alimentary Pharmabiotic Centre, Biosciences Institute, University College Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Ireland
- Corresponding author. Dept Anatomy & Neuroscience, Room 386, Western Gateway Building, University College Cork, Western Rd., Cork, Ireland.
| |
Collapse
|
108
|
Cho GH, Kim T, Son WS, Seo SH, Min SJ, Cho YS, Keum G, Jeong KS, Koh HY, Lee J, Pae AN. Synthesis and biological evaluation of aryl isoxazole derivatives as metabotropic glutamate receptor 1 antagonists: A potential treatment for neuropathic pain. Bioorg Med Chem Lett 2015; 25:1324-8. [DOI: 10.1016/j.bmcl.2015.01.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/24/2014] [Accepted: 01/19/2015] [Indexed: 10/24/2022]
|
109
|
Kobayashi S, Sato M, Kasakoshi T, Tsutsui T, Sugimoto M, Osaki M, Okada F, Igarashi K, Hiratake J, Homma T, Conrad M, Fujii J, Soga T, Bannai S, Sato H. Cystathionine is a novel substrate of cystine/glutamate transporter: implications for immune function. J Biol Chem 2015; 290:8778-88. [PMID: 25713140 DOI: 10.1074/jbc.m114.625053] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Indexed: 01/20/2023] Open
Abstract
The cystine/glutamate transporter, designated as system xc(-), is important for maintaining intracellular glutathione levels and extracellular redox balance. The substrate-specific component of system xc(-), xCT, is strongly induced by various stimuli, including oxidative stress, whereas it is constitutively expressed only in specific brain regions and immune tissues, such as the thymus and spleen. Although cystine and glutamate are the well established substrates of system xc(-) and the knockout of xCT leads to alterations of extracellular redox balance, nothing is known about other potential substrates. We thus performed a comparative metabolite analysis of tissues from xCT-deficient and wild-type mice using capillary electrophoresis time-of-flight mass spectrometry. Although most of the analyzed metabolites did not show significant alterations between xCT-deficient and wild-type mice, cystathionine emerged as being absent specifically in the thymus and spleen of xCT-deficient mice. No expression of either cystathionine β-synthase or cystathionine γ-lyase was observed in the thymus and spleen of mice. In embryonic fibroblasts derived from wild-type embryos, cystine uptake was significantly inhibited by cystathionine in a concentration-dependent manner. Wild-type cells showed an intracellular accumulation of cystathionine when incubated in cystathionine-containing buffer, which concomitantly stimulated an increased release of glutamate into the extracellular space. By contrast, none of these effects could be observed in xCT-deficient cells. Remarkably, unlike knock-out cells, wild-type cells could be rescued from cystine deprivation-induced cell death by cystathionine supplementation. We thus conclude that cystathionine is a novel physiological substrate of system xc(-) and that the accumulation of cystathionine in immune tissues is exclusively mediated by system xc(-).
Collapse
Affiliation(s)
- Sho Kobayashi
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan, the Department of Functional Genomics and Biotechnology, United Graduate School of Agricultural Sciences, Iwate University, Morioka, Iwate 020-8550, Japan, the Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Mami Sato
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| | - Takayuki Kasakoshi
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| | - Takumi Tsutsui
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| | - Masahiro Sugimoto
- the Institute of Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Mitsuhiko Osaki
- the Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago 683-8503, Japan, the Chromosome Engineering Research Center, Tottori University, Yonago, 683-8503, Japan
| | - Futoshi Okada
- the Division of Pathological Biochemistry, Tottori University Faculty of Medicine, Yonago 683-8503, Japan, the Chromosome Engineering Research Center, Tottori University, Yonago, 683-8503, Japan
| | - Kiharu Igarashi
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| | - Jun Hiratake
- the Institute for Chemical Research, Kyoto University, Uji, Kyoto 611-0011, Japan
| | - Takujiro Homma
- the Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan, and
| | - Marcus Conrad
- the Helmholtz Center Munich, German Research Center for Environmental Health, Institute of Developmental Genetics, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Junichi Fujii
- the Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan, and
| | - Tomoyoshi Soga
- the Institute of Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Shiro Bannai
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan
| | - Hideyo Sato
- From the Department of Food and Applied Life Sciences, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata 997-8555, Japan, the Department of Functional Genomics and Biotechnology, United Graduate School of Agricultural Sciences, Iwate University, Morioka, Iwate 020-8550, Japan, the Laboratory of Biochemistry and Molecular Biology, Department of Medical Technology, Faculty of Medicine, Niigata University, Chuo-ku, Niigata 951-8518, Japan
| |
Collapse
|
110
|
Monn JA, Prieto L, Taboada L, Pedregal C, Hao J, Reinhard MR, Henry SS, Goldsmith PJ, Beadle CD, Walton L, Man T, Rudyk H, Clark B, Tupper D, Baker SR, Lamas C, Montero C, Marcos A, Blanco J, Bures M, Clawson DK, Atwell S, Lu F, Wang J, Russell M, Heinz BA, Wang X, Carter JH, Xiang C, Catlow JT, Swanson S, Sanger H, Broad LM, Johnson MP, Knopp KL, Simmons RMA, Johnson BG, Shaw DB, McKinzie DL. Synthesis and Pharmacological Characterization of C4-Disubstituted Analogs of 1S,2S,5R,6S-2-Aminobicyclo[3.1.0]hexane-2,6-dicarboxylate: Identification of a Potent, Selective Metabotropic Glutamate Receptor Agonist and Determination of Agonist-Bound Human mGlu2 and mGlu3 Amino Terminal Domain Structures. J Med Chem 2015; 58:1776-94. [DOI: 10.1021/jm501612y] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- James A. Monn
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lourdes Prieto
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lorena Taboada
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Concepcion Pedregal
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Junliang Hao
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Matt R. Reinhard
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Steven S. Henry
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Paul J. Goldsmith
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Christopher D. Beadle
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lesley Walton
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Teresa Man
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Helene Rudyk
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Barry Clark
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David Tupper
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - S. Richard Baker
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Carlos Lamas
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Carlos Montero
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Alicia Marcos
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jaime Blanco
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Mark Bures
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David K. Clawson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Shane Atwell
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Frances Lu
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Jing Wang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Marijane Russell
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Beverly A. Heinz
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Xushan Wang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Joan H. Carter
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Chuanxi Xiang
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - John T. Catlow
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Steven Swanson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Helen Sanger
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Lisa M. Broad
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Michael P. Johnson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Kelly L. Knopp
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Rosa M. A. Simmons
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Bryan G. Johnson
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David B. Shaw
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - David L. McKinzie
- Discovery Chemistry
Research and Technologies, ‡Quantitative Biology, §Structural Biology, ∥Drug Disposition,
and ⊥Neuroscience
Research, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| |
Collapse
|
111
|
Yang H, Miao Q, Johnson BF, Rishel MJ, Sossi V, Dinelle K, Bénard. F, Yapp DT, Webster JM, Schaffer P. A simple route to [11C]N-Me labeling of aminosuberic acid for proof of feasibility imaging of the xC− transporter. Bioorg Med Chem Lett 2014; 24:5512-5. [DOI: 10.1016/j.bmcl.2014.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/27/2014] [Accepted: 10/01/2014] [Indexed: 01/01/2023]
|
112
|
Johnson AC, Greenwood-Van Meerveld B. Stress-induced pain: a target for the development of novel therapeutics. J Pharmacol Exp Ther 2014; 351:327-35. [PMID: 25194019 PMCID: PMC4201269 DOI: 10.1124/jpet.114.218065] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/04/2014] [Indexed: 12/12/2022] Open
Abstract
Although current therapeutics provide relief from acute pain, drugs used for treatment of chronic pain are typically less efficacious and limited by adverse side effects, including tolerance, addiction, and gastrointestinal upset. Thus, there is a significant need for novel therapies for the treatment of chronic pain. In concert with chronic pain, persistent stress facilitates pain perception and sensitizes pain pathways, leading to a feed-forward cycle promoting chronic pain disorders. Stress exacerbation of chronic pain suggests that centrally acting drugs targeting the pain- and stress-responsive brain regions represent a valid target for the development of novel therapeutics. This review provides an overview of how stress modulates spinal and central pain pathways, identifies key neurotransmitters and receptors within these pathways, and highlights their potential as novel targets for therapeutics to treat chronic pain.
Collapse
Affiliation(s)
- Anthony C Johnson
- Veterans Affairs Medical Center (B.G.-V.M.), Department of Physiology (B.G.-V.M.), and Oklahoma Center for Neuroscience (A.C.J., B.G.-V.M.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Beverley Greenwood-Van Meerveld
- Veterans Affairs Medical Center (B.G.-V.M.), Department of Physiology (B.G.-V.M.), and Oklahoma Center for Neuroscience (A.C.J., B.G.-V.M.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
113
|
Mardinoglu A, Kampf C, Asplund A, Fagerberg L, Hallström BM, Edlund K, Blüher M, Pontén F, Uhlen M, Nielsen J. Defining the human adipose tissue proteome to reveal metabolic alterations in obesity. J Proteome Res 2014; 13:5106-19. [PMID: 25219818 DOI: 10.1021/pr500586e] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
White adipose tissue (WAT) has a major role in the progression of obesity. Here, we combined data from RNA-Seq and antibody-based immunohistochemistry to describe the normal physiology of human WAT obtained from three female subjects and explored WAT-specific genes by comparing WAT to 26 other major human tissues. Using the protein evidence in WAT, we validated the content of a genome-scale metabolic model for adipocytes. We employed this high-quality model for the analysis of subcutaneous adipose tissue (SAT) gene expression data obtained from subjects included in the Swedish Obese Subjects Sib Pair study to reveal molecular differences between lean and obese individuals. We integrated SAT gene expression and plasma metabolomics data, investigated the contribution of the metabolic differences in the mitochondria of SAT to the occurrence of obesity, and eventually identified cytosolic branched-chain amino acid (BCAA) transaminase 1 as a potential target that can be used for drug development. We observed decreased glutaminolysis and alterations in the BCAAs metabolism in SAT of obese subjects compared to lean subjects. We also provided mechanistic explanations for the changes in the plasma level of BCAAs, glutamate, pyruvate, and α-ketoglutarate in obese subjects. Finally, we validated a subset of our model-based predictions in 20 SAT samples obtained from 10 lean and 10 obese male and female subjects.
Collapse
Affiliation(s)
- Adil Mardinoglu
- Department of Chemical and Biological Engineering, Chalmers University of Technology , 412 96 Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Mercier MS, Lodge D. Group III metabotropic glutamate receptors: pharmacology, physiology and therapeutic potential. Neurochem Res 2014; 39:1876-94. [PMID: 25146900 DOI: 10.1007/s11064-014-1415-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/08/2014] [Accepted: 08/11/2014] [Indexed: 01/14/2023]
Abstract
Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), exerts neuromodulatory actions via the activation of metabotropic glutamate (mGlu) receptors. There are eight known mGlu receptor subtypes (mGlu1-8), which are widely expressed throughout the brain, and are divided into three groups (I-III), based on signalling pathways and pharmacological profiles. Group III mGlu receptors (mGlu4/6/7/8) are primarily, although not exclusively, localised on presynaptic terminals, where they act as both auto- and hetero-receptors, inhibiting the release of neurotransmitter. Until recently, our understanding of the role of individual group III mGlu receptor subtypes was hindered by a lack of subtype-selective pharmacological tools. Recent advances in the development of both orthosteric and allosteric group III-targeting compounds, however, have prompted detailed investigations into the possible functional role of these receptors within the CNS, and revealed their involvement in a number of pathological conditions, such as epilepsy, anxiety and Parkinson's disease. The heterogeneous expression of group III mGlu receptor subtypes throughout the brain, as well as their distinct distribution at glutamatergic and GABAergic synapses, makes them ideal targets for therapeutic intervention. This review summarises the advances in subtype-selective pharmacology, and discusses the individual roles of group III mGlu receptors in physiology, and their potential involvement in disease.
Collapse
Affiliation(s)
- Marion S Mercier
- Centre for Synaptic Plasticity, School of Physiology and Pharmacology, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK,
| | | |
Collapse
|
115
|
Sedic M, Gethings LA, Vissers JPC, Shockcor JP, McDonald S, Vasieva O, Lemac M, Langridge JI, Batinić D, Pavelić SK. Label-free mass spectrometric profiling of urinary proteins and metabolites from paediatric idiopathic nephrotic syndrome. Biochem Biophys Res Commun 2014; 452:21-6. [PMID: 25150443 DOI: 10.1016/j.bbrc.2014.08.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 08/04/2014] [Indexed: 01/20/2023]
Abstract
Idiopathic nephrotic syndrome (INS) is caused by renal diseases that increase the permeability of the glomerular filtration barrier without evidence of a specific systemic cause. The aim of the present work was to reveal inherent molecular features of INS in children using combined urinary proteomics and metabolomics profiling. In this study, label-free mass spectrometric analysis of urinary proteins and small molecule metabolites was carried out in 12 patients with INS versus 12 sex- and age-matched control subjects with normal renal function. Integration and biological interpretation of obtained results were carried out by Ingenuity IPA software. Validation of obtained proteomics data was carried out by Western blot method. Proteomics data have been deposited to the ProteomeXchange Consortium with the data set identifier PXD000765. This study indicates for the first time that paediatric INS is associated with up-regulation of afamin, hydroxyphenylacetate and uridine, and concomitant down-regulation in glutamine and phenylalanine levels, and many of these molecular species were previously shown to be involved in oxidative stress. Further studies in larger patient population are underway to investigate the role of oxidative stress in renal injury in paediatric INS.
Collapse
Affiliation(s)
- Mirela Sedic
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Lee A Gethings
- Waters Corporation, MS Technologies, Manchester, United Kingdom
| | | | | | | | - Olga Vasieva
- Department of Functional and Comparative Genomics, Institute of Integrative Biology, University of Liverpool, United Kingdom
| | - Maja Lemac
- Division of Nephrology, Department of Pediatrics, Zagreb University Hospital Centre, Zagreb, Croatia
| | | | - Danica Batinić
- Division of Nephrology, Department of Pediatrics, Zagreb University Hospital Centre, Zagreb, Croatia
| | | |
Collapse
|
116
|
Pokusa M, Prokopova B, Hlavacova N, Makatsori A, Jezova D. Effect of blockade of mGluR5 on stress hormone release and its gene expression in the adrenal gland. Can J Physiol Pharmacol 2014; 92:686-92. [PMID: 25019607 DOI: 10.1139/cjpp-2014-0030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to verify the presence of metabotropic glutamate receptor subtype 5 (mGluR5) in the adrenal gland of male rats of 2 different strains, and to test the hypothesis that treatment with mGluR5 antagonist 2-methyl-6-(phenylethynyl)-pyridine (MPEP) affects hormone release and adrenal gene expression of mGluR5 under conditions of stress. The results clearly show the gene expression of mGluR5 in the adrenal gland in both the adrenal cortex and medulla. Treatment with the glutamate release inhibitor riluzole (4 mg·(kg body mass)(-1)·day(-1) for 2 weeks) failed to modify mRNA levels of either the mGluR5 or NR1 subunit of the NMDA receptor in the adrenal glands, as measured by real-time PCR. Blockade of mGluR5 with MPEP (1 mg·kg(-1) for 4 days) increased corticosterone but not catecholamine release during restraint stress (20 min). Treatment with MPEP had no effect on mRNA levels coding for steroidogenic factors StAR and SF-1, and decreased mGluR5 gene expression in the adrenal gland. In conclusion, mGluR5 is not likely to play a significant role in stress-induced catecholamine release. Pharmacological blockade of mGluR5 has a modest influence on the hypothalamic-pituitary-adrenocortical axis, as reflected in adrenal hypertrophy and increased corticosterone concentrations.
Collapse
Affiliation(s)
- Michal Pokusa
- a Laboratory of Pharmacological Neuroendocrinology, Institute of Experimental Endocrinology, Slovak Academy of Sciences, Vlarska 3, Bratislava 83306, Slovakia
| | | | | | | | | |
Collapse
|
117
|
Increased response to glutamate in small diameter dorsal root ganglion neurons after sciatic nerve injury. PLoS One 2014; 9:e95491. [PMID: 24748330 PMCID: PMC3991716 DOI: 10.1371/journal.pone.0095491] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Accepted: 03/26/2014] [Indexed: 12/24/2022] Open
Abstract
Glutamate in the peripheral nervous system is involved in neuropathic pain, yet we know little how nerve injury alters responses to this neurotransmitter in primary sensory neurons. We recorded neuronal responses from the ex-vivo preparations of the dorsal root ganglia (DRG) one week following a chronic constriction injury (CCI) of the sciatic nerve in adult rats. We found that small diameter DRG neurons (<30 µm) exhibited increased excitability that was associated with decreased membrane threshold and rheobase, whereas responses in large diameter neurons (>30 µm) were unaffected. Puff application of either glutamate, or the selective ionotropic glutamate receptor agonists alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and kainic acid (KA), or the group I metabotropic receptor (mGluR) agonist (S)-3,5-dihydroxyphenylglycine (DHPG), induced larger inward currents in CCI DRGs compared to those from uninjured rats. N-methyl-D-aspartate (NMDA)-induced currents were unchanged. In addition to larger inward currents following CCI, a greater number of neurons responded to glutamate, AMPA, NMDA, and DHPG, but not to KA. Western blot analysis of the DRGs revealed that CCI resulted in a 35% increase in GluA1 and a 60% decrease in GluA2, the AMPA receptor subunits, compared to uninjured controls. mGluR1 receptor expression increased by 60% in the membrane fraction, whereas mGluR5 receptor subunit expression remained unchanged after CCI. These results show that following nerve injury, small diameter DRG neurons, many of which are nociceptive, have increased excitability and an increased response to glutamate that is associated with changes in receptor expression at the neuronal membrane. Our findings provide further evidence that glutamatergic transmission in the periphery plays a role in nociception.
Collapse
|
118
|
Golubeva AV, Zhdanov AV, Mallel G, Dinan TG, Cryan JF. The mouse cyclophosphamide model of bladder pain syndrome: tissue characterization, immune profiling, and relationship to metabotropic glutamate receptors. Physiol Rep 2014; 2:e00260. [PMID: 24760514 PMCID: PMC4002240 DOI: 10.1002/phy2.260] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Accepted: 02/17/2014] [Indexed: 01/08/2023] Open
Abstract
Abstract Painful bladder syndrome/Interstitial cystitis (PBS/IC) is a chronic disorder characterized clinically by recurring episodes of pelvic pain and increased urination frequency, significantly impairing patients' quality of life. Despite this, there is an unmet medical need in terms of effective diagnostics and treatment. Animal models are crucial in this endeavor. Systemic chronic administration of cyclophosphamide (CYP) in mice has been proposed as a relevant preclinical model of chronic bladder pain. However, molecular mechanisms underlying the pathogenesis of this model are lacking. Here, we show that mice, subjected to repetitive systemic injections of CYP, developed mild inflammatory response in bladder tissue characterized by submucosal edema, moderate increase in proinflammatory cytokine gene expression, and mastocytosis. No signs of massive inflammatory infiltrate, tissue hemorrhages, mucosal ulcerations and urothelium loss were observed. Instead, CYP treatment induced urothelium hyperplasia, accompanied by activation of proliferative signaling cascades, and a decrease in the expression of urothelium-specific markers. Metabotropic glutamate (mGlu) receptors have been implicated in chronic pain disorders. CYP administration induced differential changes in mGlu receptors mRNA levels in bladder tissue, without affecting gene expression at spinal cord level, pointing to the potential link between peripheral mGlu receptors and inflammation-induced bladder malfunction and hyperalgesia. Taken together, these data indicate that chronic CYP treatment in mice is a model of PBS mostly relevant to the major, nonulcerative subtype of the syndrome, characterized by a relatively unaltered mucosa and a sparse inflammatory response. This model can help to elucidate the pathogenetic mechanisms of the disease.
Collapse
Affiliation(s)
- Anna V. Golubeva
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | - Giuseppe Mallel
- Pathology Unit, Department of Clinical and Molecular Medicine, S. Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Timothy G. Dinan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
- Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F. Cryan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
119
|
de Lartigue G. Putative roles of neuropeptides in vagal afferent signaling. Physiol Behav 2014; 136:155-69. [PMID: 24650553 DOI: 10.1016/j.physbeh.2014.03.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/23/2014] [Accepted: 03/10/2014] [Indexed: 02/06/2023]
Abstract
The vagus nerve is a major pathway by which information is communicated between the brain and peripheral organs. Sensory neurons of the vagus are located in the nodose ganglia. These vagal afferent neurons innervate the heart, the lung and the gastrointestinal tract, and convey information about peripheral signals to the brain important in the control of cardiovascular tone, respiratory tone, and satiation, respectively. Glutamate is thought to be the primary neurotransmitter involved in conveying all of this information to the brain. It remains unclear how a single neurotransmitter can regulate such an extensive list of physiological functions from a wide range of visceral sites. Many neurotransmitters have been identified in vagal afferent neurons and have been suggested to modulate the physiological functions of glutamate. Specifically, the anorectic peptide transmitters, cocaine and amphetamine regulated transcript (CART) and the orexigenic peptide transmitters, melanin concentrating hormone (MCH) are differentially regulated in vagal afferent neurons and have opposing effects on food intake. Using these two peptides as a model, this review will discuss the potential role of peptide transmitters in providing a more precise and refined modulatory control of the broad physiological functions of glutamate, especially in relation to the control of feeding.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Dept Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA.
| |
Collapse
|
120
|
The role of glutamate and its receptors in multiple sclerosis. J Neural Transm (Vienna) 2014; 121:945-55. [PMID: 24633998 DOI: 10.1007/s00702-014-1188-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 02/27/2014] [Indexed: 12/18/2022]
Abstract
Glutamate is an excitatory neurotransmitter of the central nervous system, which has a central role in a complex communication network established between neurons, astrocytes, oligodendrocytes, and microglia. Multiple abnormal triggers such as energy deficiency, oxidative stress, mitochondrial dysfunction, and calcium overload can lead to abnormalities in glutamate signaling. Thus, the disturbance of glutamate homeostasis could affect practically all physiological functions and interactions of brain cells, leading to excitotoxicity. Excitotoxicity is the pathological process by which nerve cells are damaged or killed by excessive stimulation by glutamate. Although neuron degeneration and death are the ultimate consequences of multiple sclerosis (MS), it is now widely accepted that alterations in the function of surrounding glial cells are key features in the progression of the disease. The present knowledge raise the possibility that the modulation of glutamate release and transport, as well as receptors blockade or glutamate metabolism modulation, might be relevant targets for the development of future therapeutic interventions in MS.
Collapse
|
121
|
Webster JM, Morton CA, Johnson BF, Yang H, Rishel MJ, Lee BD, Miao Q, Pabba C, Yapp DT, Schaffer P. Functional imaging of oxidative stress with a novel PET imaging agent, 18F-5-fluoro-L-aminosuberic acid. J Nucl Med 2014; 55:657-64. [PMID: 24578242 DOI: 10.2967/jnumed.113.126664] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED Glutathione is the predominant endogenous cellular antioxidant, playing a critical role in the cellular defensive response to oxidative stress by neutralizing free radicals and reactive oxygen species. With cysteine as the rate-limiting substrate in glutathione biosynthesis, the cystine/glutamate transporter (system xc(-)) represents a potentially attractive PET biomarker to enable in vivo quantification of xc(-) activity in response to oxidative stress associated with disease. We have developed a system xc(-) substrate that incorporates characteristics of both natural substrates, L-cystine and L-glutamate (L-Glu). L-aminosuberic acid (L-ASu) has been identified as a more efficient system xc(-) substrate than L-Glu, leading to an assessment of a series of anionic amino acids as prospective PET tracers. Herein, we report the synthesis and in vitro and in vivo validation of a lead candidate, (18)F-5-fluoro-aminosuberic acid ((18)F-FASu), as a PET tracer for functional imaging of a cellular response to oxidative stress with remarkable tumor uptake and retention. METHODS (18)F-FASu was identified as a potential PET tracer based on an in vitro screening of compounds similar to L-cystine and L-Glu. Affinity toward system xc(-) was determined via in vitro uptake and inhibition studies using oxidative stress-induced EL4 and SKOV-3 cells. In vivo biodistribution and PET imaging studies were performed in mice bearing xenograft tumors (EL4 and SKOV-3). RESULTS In vitro assay results determined that L-ASu inhibited system xc(-) as well as or better than L-Glu. The direct comparison of uptake of tritiated compounds demonstrated more efficient system xc(-) uptake of L-ASu than L-Glu. Radiosynthesis of (18)F-FASu allowed the validation of uptake for the fluorine-bearing derivative in vitro. Evaluation in vivo demonstrated primarily renal clearance and uptake of approximately 8 percentage injected dose per gram in SKOV-3 tumors, with tumor-to-blood and tumor-to-muscle ratios of approximately 12 and approximately 28, respectively. (18)F-FASu uptake was approximately 5 times greater than (18)F-FDG uptake in SKOV-3 tumors. Dynamic PET imaging demonstrated uptake in EL4 tumor xenografts of approximately 6 percentage injected dose per gram and good tumor retention for at least 2 h after injection. CONCLUSION (18)F-FASu is a potentially useful metabolic tracer for PET imaging of a functional cellular response to oxidative stress. (18)F-FASu may provide more sensitive detection than (18)F-FDG in certain tumors.
Collapse
Affiliation(s)
- Jack M Webster
- Diagnostics and Biomedical Technologies, GE Global Research, Niskayuna, New York
| | | | | | | | | | | | | | | | | | | |
Collapse
|
122
|
Felizola SJA, Nakamura Y, Satoh F, Morimoto R, Kikuchi K, Nakamura T, Hozawa A, Wang L, Onodera Y, Ise K, McNamara KM, Midorikawa S, Suzuki S, Sasano H. Glutamate receptors and the regulation of steroidogenesis in the human adrenal gland: the metabotropic pathway. Mol Cell Endocrinol 2014; 382:170-177. [PMID: 24080311 DOI: 10.1016/j.mce.2013.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Revised: 09/18/2013] [Accepted: 09/20/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND l-glutamate is a major excitatory neurotransmitter in the mammalian brain. Glutamate receptors have been reported in the rat adrenal cortex and in human aldosterone-producing adenomas (APA). However, details regarding the expression levels and functions of these receptors in human adrenocortical tissues remain unknown. METHODS The mRNA levels of glutamate receptors were evaluated by qPCR in: 12 normal adrenal cortex (NAC), 11 APA, and 12 cortisol-producing adenoma (CPA) tissues. Protein localization was evaluated by immunohistochemistry for 15 NAC, 5 idiopathic hyperaldosteronism cases, 15 APA and 15 CPA. H295R cells were treated with angiotensin-II or forskolin alone or combined with the GRM2/3 agonist LY354740. RESULTS The level of GRM3 mRNA was higher in APA than in CPA (P=0.0086) or NAC (P=0.0022). GRM1, IGLUR2, and IGLUR3 were also detected in adrenocortical tissues. When added to angiotensin-II/forskolin treatments, LY354740 decreased aldosterone and cortisol production in H295R cells. CONCLUSIONS GRM3 is considered to regulate steroidogenesis in adrenocortical tissues.
Collapse
Affiliation(s)
- Saulo J A Felizola
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasuhiro Nakamura
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Fumitoshi Satoh
- Division of Nephrology and Hypertension, Tohoku University Hospital, Sendai, Japan
| | - Ryo Morimoto
- Division of Nephrology and Hypertension, Tohoku University Hospital, Sendai, Japan
| | - Kumi Kikuchi
- Division of Nephrology and Hypertension, Tohoku University Hospital, Sendai, Japan
| | - Tomohiro Nakamura
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Atsushi Hozawa
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Lin Wang
- Department of Physiology, Harbin Medical University Daqing Branch, Daqing, China
| | - Yoshiaki Onodera
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazue Ise
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keely M McNamara
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sanae Midorikawa
- Department of Radiation Health Management, Fukushima Medical University, Fukushima, Japan
| | - Shinichi Suzuki
- Department of Organ Regulatory Surgery, Fukushima Medical University, Fukushima, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
123
|
Zhou Y, Waanders LF, Holmseth S, Guo C, Berger UV, Li Y, Lehre AC, Lehre KP, Danbolt NC. Proteome analysis and conditional deletion of the EAAT2 glutamate transporter provide evidence against a role of EAAT2 in pancreatic insulin secretion in mice. J Biol Chem 2013; 289:1329-44. [PMID: 24280215 DOI: 10.1074/jbc.m113.529065] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Islet function is incompletely understood in part because key steps in glutamate handling remain undetermined. The glutamate (excitatory amino acid) transporter 2 (EAAT2; Slc1a2) has been hypothesized to (a) provide islet cells with glutamate, (b) protect islet cells against high extracellular glutamate concentrations, (c) mediate glutamate release, or (d) control the pH inside insulin secretory granules. Here we floxed the EAAT2 gene to produce the first conditional EAAT2 knock-out mice. Crossing with Nestin-cyclization recombinase (Cre) eliminated EAAT2 from the brain, resulting in epilepsy and premature death, confirming the importance of EAAT2 for brain function and validating the genetic construction. Crossing with insulin-Cre lines (RIP-Cre and IPF1-Cre) to obtain pancreas-selective deletion did not appear to affect survival, growth, glucose tolerance, or β-cell number. We found (using TaqMan RT-PCR, immunoblotting, immunocytochemistry, and proteome analysis) that the EAAT2 levels were too low to support any of the four hypothesized functions. The proteome analysis detected more than 7,000 islet proteins of which more than 100 were transporters. Although mitochondrial glutamate transporters and transporters for neutral amino acids were present at high levels, all other transporters with known ability to transport glutamate were strikingly absent. Glutamate-metabolizing enzymes were abundant. The level of glutamine synthetase was 2 orders of magnitude higher than that of glutaminase. Taken together this suggests that the uptake of glutamate by islets from the extracellular fluid is insignificant and that glutamate is intracellularly produced. Glutamine synthetase may be more important for islets than assumed previously.
Collapse
Affiliation(s)
- Yun Zhou
- From The Neurotransporter Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Notartomaso S, Zappulla C, Biagioni F, Cannella M, Bucci D, Mascio G, Scarselli P, Fazio F, Weisz F, Lionetto L, Simmaco M, Gradini R, Battaglia G, Signore M, Puliti A, Nicoletti F. Pharmacological enhancement of mGlu1 metabotropic glutamate receptors causes a prolonged symptomatic benefit in a mouse model of spinocerebellar ataxia type 1. Mol Brain 2013; 6:48. [PMID: 24252411 PMCID: PMC4225515 DOI: 10.1186/1756-6606-6-48] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 10/28/2013] [Indexed: 11/10/2022] Open
Abstract
Background Spinocerebellar ataxia type 1 (SCA1) is a genetic disorder characterized by severe ataxia associated with progressive loss of cerebellar Purkinje cells. The mGlu1 metabotropic glutamate receptor plays a key role in mechanisms of activity-dependent synaptic plasticity in the cerebellum, and its dysfunction is linked to the pathophysiology of motor symptoms associated with SCA1. We used SCA1 heterozygous transgenic mice (Q154/Q2) as a model for testing the hypothesis that drugs that enhance mGlu1 receptor function may be good candidates for the medical treatment of SCA1. Results Symptomatic 30-week old SCA1 mice showed reduced mGlu1 receptor mRNA and protein levels in the cerebellum. Interestingly, these mice also showed an intense expression of mGlu5 receptors in cerebellar Purkinje cells, which normally lack these receptors. Systemic treatment of SCA1 mice with the mGlu1 receptor positive allosteric modulator (PAM), Ro0711401 (10 mg/kg, s.c.), caused a prolonged improvement of motor performance on the rotarod and the paw-print tests. A single injection of Ro0711401 improved motor symptoms for several days, and no tolerance developed to the drug. In contrast, the mGlu5 receptor PAM, VU0360172 (10 mg/kg, s.c.), caused only a short-lasting improvement of motor symptoms, whereas the mGlu1 receptor antagonist, JNJ16259685 (2.5 mg/kg, i.p.), further impaired motor performance in SCA1 mice. The prolonged symptomatic benefit caused by Ro0711401 outlasted the time of drug clearance from the cerebellum, and was associated with neuroadaptive changes in the cerebellum, such as a striking reduction of the ectopically expressed mGlu5 receptors in Purkinje cells, increases in levels of total and Ser880-phosphorylated GluA2 subunit of AMPA receptors, and changes in the length of spines in the distal dendrites of Purkinje cells. Conclusions These data demonstrate that pharmacological enhancement of mGlu1 receptors causes a robust and sustained motor improvement in SCA1 mice, and lay the groundwork for the development of mGlu1 receptor PAMs as novel “cerebellum-specific”, effective, and safe symptomatic drugs for the treatment of SCA1 in humans.
Collapse
|
125
|
Plasma amino acids changes in complex regional pain syndrome. PAIN RESEARCH AND TREATMENT 2013; 2013:742407. [PMID: 24303215 PMCID: PMC3835366 DOI: 10.1155/2013/742407] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/18/2013] [Accepted: 09/22/2013] [Indexed: 11/18/2022]
Abstract
Complex regional pain syndrome (CRPS) is a severe chronic pain condition that most often develops following trauma. Blood samples were collected from 220 individuals, 160 CRPS subjects, and 60 healthy pain-free controls. Plasma amino acid levels were compared and contrasted between groups. L-Aspartate, L-glutamate, and L-ornithine were significantly increased, whereas L-tryptophan and L-arginine were significantly decreased in CRPS subjects as compared to controls. In addition, the L-kynurenine to L-tryptophan ratio demonstrated a significant increase, whereas the global arginine bioavailability ratio (GABR) was significantly decreased in the CRPS subjects. The CRPS subjects demonstrated a significant correlation between overall pain and the plasma levels of L-glutamate and the L-kynurenine to L-tryptophan ratio. CRPS subjects also showed a correlation between the decrease in plasma L-tryptophan and disease duration. This study shows that CRPS subjects exhibit significant changes in plasma levels of amino acids involved in glutamate receptor activation and in amino acids associated with immune function as compared to healthy pain-free controls. A better understanding of the role plasma amino acids play in the pathophysiology of CRPS may lead to novel treatments for this crippling condition.
Collapse
|
126
|
Iacovelli L, Felicioni M, Nisticò R, Nicoletti F, De Blasi A. Selective regulation of recombinantly expressed mGlu7 metabotropic glutamate receptors by G protein-coupled receptor kinases and arrestins. Neuropharmacology 2013; 77:303-12. [PMID: 24148810 DOI: 10.1016/j.neuropharm.2013.10.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/24/2013] [Accepted: 10/07/2013] [Indexed: 12/24/2022]
Abstract
mGlu7 receptors are coupled to Gi/Go-proteins and activate multiple transduction pathways, including inhibition of adenylyl cyclase activity and stimulation of ERK1/2 and JNK pathways. mGlu7 receptors play an important role in cognition and emotion and are involved in stress-related disorders such as anxiety and depression and in susceptibility to convulsive seizures. In spite of these potential clinical implications, little is known on the mechanisms that regulate mGlu7-receptor signaling. Here we show that mGlu7 receptor-dependent signaling pathways were regulated in a complementary manner by different GRK subtypes, with GRK4 affecting the adenylyl cyclase and the JNK pathways, and GRK2 selectively affecting the ERK1/2 pathway. Additionally we found that the two isoforms of non-visual arrestins, i.e. β-arrestin1 and β-arrestin2, exerted opposite effects on mGlu7-receptor signaling, with β-arrestin1 positively modulating ERK1/2 and inhibiting JNK, and β-arrestin2 doing the opposite. This represents a remarkable example of "reciprocal regulation" of receptor signaling by the two isoforms of β-arrestin. Finally we found that β-arrestin1 amplified mGlu7 receptor-dependent ERK1/2 activation in response to L-AP4 (an orthosteric agonist), but not in response to AMN082 (an atypical mGlu7-receptor allosteric agonist). The different effect of β-arrestin1 on L-AP4- and AMN082-stimulated ERK1/2 phosphorylation is in line with the emerging concept of β-arrestin-biased agonists. The present study may open new perspectives in elucidating the physio-pathological roles of the mGlu7 receptor and may provide new insights for the possibility to develop specific (biased) agonists that can selectively activate different signaling pathways.
Collapse
Affiliation(s)
- L Iacovelli
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy.
| | - M Felicioni
- IRCSS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - R Nisticò
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy; IRCSS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - F Nicoletti
- Department of Physiology and Pharmacology "Vittorio Erspamer", University of Rome "Sapienza", P.le Aldo Moro, 5, 00185 Rome, Italy; I.N.M. Neuromed, Località Camerelle, Pozzilli, Italy
| | - A De Blasi
- Dept. of Molecular Medicine, University of Rome "Sapienza", V.le Regina Elena 291, 00185 Rome, Italy
| |
Collapse
|
127
|
Willard SS, Koochekpour S. Glutamate, glutamate receptors, and downstream signaling pathways. Int J Biol Sci 2013; 9:948-59. [PMID: 24155668 PMCID: PMC3805900 DOI: 10.7150/ijbs.6426] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/11/2013] [Indexed: 12/23/2022] Open
Abstract
Glutamate is a nonessential amino acid, a major bioenergetic substrate for proliferating normal and neoplastic cells, and an excitatory neurotransmitter that is actively involved in biosynthetic, bioenergetic, metabolic, and oncogenic signaling pathways. Glutamate signaling activates a family of receptors consisting of metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs), both of which have been implicated in chronic disabling brain disorders such as Schizophrenia and neurodegenerative diseases like Alzheimer's, Parkinson's, and multiple sclerosis. In this review, we discuss the structural and functional relationship of mGluRs and iGluRs and their downstream signaling pathways. The three groups of mGluRs, the associated second messenger systems, and subsequent activation of PI3K/Akt, MAPK, NFkB, PLC, and Ca/CaM signaling systems will be discussed in detail. The current state of human mGluR1a as one of the most important isoforms of Group I-mGluRs will be highlighted. The lack of studies on the human orthologues of mGluRs family will be outlined. We conclude that upon further study, human glutamate-initiated signaling pathways may provide novel therapeutic opportunities for a variety of non-malignant and malignant human diseases.
Collapse
Affiliation(s)
- Stacey S Willard
- Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY, USA
| | | |
Collapse
|
128
|
Willard SS, Koochekpour S. Glutamate signaling in benign and malignant disorders: current status, future perspectives, and therapeutic implications. Int J Biol Sci 2013; 9:728-42. [PMID: 23983606 PMCID: PMC3753409 DOI: 10.7150/ijbs.6475] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/15/2013] [Indexed: 11/05/2022] Open
Abstract
Glutamate, a nonessential amino acid, is the major excitatory neurotransmitter in the central nervous system. As such, glutamate has been shown to play a role in not only neural processes, such as learning and memory, but also in bioenergetics, biosynthetic and metabolic oncogenic pathways. Glutamate has been the target of intense investigation for its involvement not only in the pathogenesis of benign neurodegenerative diseases (NDDs) such as Parkinson's disease, Alzheimer's disease, schizophrenia, multiple sclerosis, and amyotropic lateral sclerosis (ALS), but also in carcinogenesis and progression of malignant diseases. In addition to its intracellular activities, glutamate in secreted form is a phylogenetically conserved cell signaling molecule. Glutamate binding activates multiple major receptor families including the metabotropic glutamate receptors (mGluRs) and ionotropic glutamate receptors (iGluRs), both of which have been implicated in various signaling pathways in cancer. Inhibition of extracellular glutamate release or glutamate receptor activation via competitive or non-competitive antagonists decreases growth, migration and invasion and induces apoptosis in breast cancer, melanoma, glioma and prostate cancer cells. In this review, we discuss the current state of glutamate signaling research as it relates to benign and malignant diseases. In addition, we provide a synopsis of clinical trials using glutamate antagonists for the treatment of NDD and malignant diseases. We conclude that in addition to its potential role as a metabolic biomarker, glutamate receptors and glutamate-initiated signaling pathways may provide novel therapeutic opportunities for cancer.
Collapse
Affiliation(s)
- Stacey S Willard
- Departments of Cancer Genetics and Urology, Center for Genetics and Pharmacology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
129
|
Eisenkraft A, Falk A, Finkelstein A. The role of glutamate and the immune system in organophosphate-induced CNS damage. Neurotox Res 2013; 24:265-79. [PMID: 23532600 DOI: 10.1007/s12640-013-9388-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 03/10/2013] [Accepted: 03/15/2013] [Indexed: 12/12/2022]
Abstract
Organophosphate (OP) poisoning is associated with long-lasting neurological damage, which is attributed mainly to the excessive levels of glutamate caused by the intoxication. Glutamate toxicity, however, is not specific to OP poisoning, and is linked to propagation of damage in both acute and chronic neurodegenerative conditions in the central nervous system (CNS). In addition to acute excitotoxic effects of glutamate, there is now a growing amount of evidence of its intricate immunomodulatory effects in the brain, involving both the innate and the adaptive immune systems. Moreover, it was demonstrated that immunomodulatory treatments, aimed at regulating the interaction between the resident immune cells of the brain (microglia) and the peripheral immune system, can support buffering of excessive levels of glutamate and restoration of the homeostasis. In this review, we will discuss the role of glutamate as an excitotoxic agent in the acute phase of OP poisoning, and the possible functions it may have as both a neuroprotectant and an immunomodulator in the sub-acute and chronic phases of OP poisoning. In addition, we will describe the novel immune-based neuroprotective strategies aimed at counteracting the long-term neurodegenerative effects of glutamate in the CNS.
Collapse
|
130
|
Tonack S, Tang C, Offermanns S. Endogenous metabolites as ligands for G protein-coupled receptors modulating risk factors for metabolic and cardiovascular disease. Am J Physiol Heart Circ Physiol 2012; 304:H501-13. [PMID: 23241321 DOI: 10.1152/ajpheart.00641.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During the last decade, several G protein-coupled receptors activated by endogenous metabolites have been described. These receptors respond to fatty acids, mono- and disaccharides, amino acids, or various intermediates and products of metabolism, including ketone bodies, lactate, succinate, or bile acids. Receptors of endogenous metabolites are expressed in taste cells, the gastrointestinal tract, adipose tissue, endocrine glands, immune cells, or the kidney and are therefore in a position to sense food intake in the gastrointestinal tract or to link metabolite levels to the appropriate responses of metabolic organs. Some of the receptors appear to provide a link between metabolic and neuronal or immune functions. Given that many of these metabolic processes are dysregulated under pathological conditions, including diabetes, dyslipidemia, and obesity, receptors of endogenous metabolites have also been recognized as potential drug targets to prevent and/or treat metabolic and cardiovascular diseases. This review describes G protein-coupled receptors activated by endogenous metabolites and summarizes their physiological, pathophysiological, and potential pharmacological roles.
Collapse
Affiliation(s)
- Sarah Tonack
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | | | | |
Collapse
|
131
|
Podocytes: A new player for glutamate signaling. Int J Biochem Cell Biol 2012; 44:2272-7. [DOI: 10.1016/j.biocel.2012.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 07/20/2012] [Accepted: 09/16/2012] [Indexed: 11/24/2022]
|
132
|
Morrow JA, Gilfillan R, Neale SA. Glutamatergic Approaches for the Treatment of Schizophrenia. DRUG DISCOVERY FOR PSYCHIATRIC DISORDERS 2012. [DOI: 10.1039/9781849734943-00056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system and plays a key role in most aspects of normal brain function including cognition, learning and memory. Dysfunction of glutamatergic neurotransmission has been implicated in a number of neurological and psychiatric disorders with a growing body of evidence suggesting that hypofunction of glutamatergic neurotransmission via the N-methyl-d-aspartate (NMDA) receptor plays an important role in the pathophysiology of schizophrenia. It thus follows that potentiation of NMDA receptor function via pharmacological manipulation may provide therapeutic utility for the treatment of schizophrenia and a number of different approaches are currently being pursued by the pharmaceutical industry with this aim in mind. These include strategies that target the glycine/d-serine site of the NMDA receptor (glycine transporter GlyT1, d-serine transporter ASC-1 and d-amino acid oxidase (DAAO) inhibitors) together with those aimed at enhancing glutamatergic neurotransmission via modulation of AMPA receptor and metabotropic glutamate receptor function. Such efforts are now beginning to bear fruit with compounds such as the GlyT1 inhibitor RG1678 and mGlu2 agonist LY2140023 proving to have clinical meaningful effects in phase II clinical trials. While more studies are required to confirm long-term efficacy, functional outcome and safety in schizophrenic agents, these agents hold real promise for addressing unmet medical needs, in particular refractory negative and cognitive symptoms, not currently addressed by existing antipsychotic agents.
Collapse
Affiliation(s)
- John A. Morrow
- Neuroscience and Ophthalmology, Merck Research Laboratories 2015 Galloping Hill Road, Kenilworth, New Jersey 07033 USA
| | - Robert Gilfillan
- Discovery Chemistry, Merck Research Laboratories 770 Sumneytown Pike, West Point, Pennsylvania 19486 USA
| | - Stuart A. Neale
- Neurexpert Ltd Ground Floor, 2 Woodberry Grove, North Finchley, London, N12 0DR UK
| |
Collapse
|
133
|
Julio-Pieper M, O'Connor RM, Dinan TG, Cryan JF. Regulation of the brain-gut axis by group III metabotropic glutamate receptors. Eur J Pharmacol 2012; 698:19-30. [PMID: 23123053 DOI: 10.1016/j.ejphar.2012.10.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/11/2012] [Accepted: 10/22/2012] [Indexed: 01/14/2023]
Abstract
L-glutamate is produced by a great variety of peripheral tissues in both health and disease. Like other components of the glutamatergic system, metabotropic glutamate (mGlu) receptors also have a widespread distribution outside the central nervous system (CNS). In particular, group III mGlu receptors have been recently found in human stomach and colon revealing an extraordinary potential for these receptors in the treatment of peripheral disorders, including gastrointestinal dysfunction. The significance of these findings is that pharmacological tools originally designed for mGlu receptors in the CNS may also be directed towards new disease targets in the periphery. Targeting mGlu receptors can also be beneficial in the treatment of disorders involving central components together with gastrointestinal dysfunction, such as irritable bowel syndrome, which can be co-morbid with anxiety and depression. Conversely, the development of more specific therapeutic approaches for mGlu ligands both centrally as in the gut will depend on the elucidation of tissue-specific elements in mGlu receptor signalling.
Collapse
Affiliation(s)
- Marcela Julio-Pieper
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Av Universidad 330, Curauma, Valparaíso, Chile.
| | | | | | | |
Collapse
|
134
|
Discovery of 1H-pyrrolo[2,3-c]pyridine-7-carboxamides as novel, allosteric mGluR5 antagonists. Bioorg Med Chem Lett 2012; 22:6454-9. [DOI: 10.1016/j.bmcl.2012.08.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 08/10/2012] [Accepted: 08/13/2012] [Indexed: 11/21/2022]
|
135
|
Davalli AM, Perego C, Folli FB. The potential role of glutamate in the current diabetes epidemic. Acta Diabetol 2012; 49:167-83. [PMID: 22218826 DOI: 10.1007/s00592-011-0364-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/19/2011] [Indexed: 12/27/2022]
Abstract
In the present article, we propose the perspective that abnormal glutamate homeostasis might contribute to diabetes pathogenesis. Previous reports and our recent data indicate that chronically high extracellular glutamate levels exert direct and indirect effects that might participate in the progressive loss of β-cells occurring in both T1D and T2D. In addition, abnormal glutamate homeostasis may impact all the three accelerators of the "accelerator hypothesis" and could partially explain the rising frequency of T1D and T2D.
Collapse
Affiliation(s)
- Alberto M Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, San Raffaele Scientific Institute, 20132, Milan, Italy.
| | | | | |
Collapse
|
136
|
Dobi A, Sartori SB, Busti D, Van der Putten H, Singewald N, Shigemoto R, Ferraguti F. Neural substrates for the distinct effects of presynaptic group III metabotropic glutamate receptors on extinction of contextual fear conditioning in mice. Neuropharmacology 2012; 66:274-89. [PMID: 22643400 PMCID: PMC3557389 DOI: 10.1016/j.neuropharm.2012.05.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 05/09/2012] [Accepted: 05/17/2012] [Indexed: 12/22/2022]
Abstract
The group III metabotropic glutamate (mGlu) receptors mGlu7 and mGlu8 are receiving increased attention as potential novel therapeutic targets for anxiety disorders. The effects mediated by these receptors appear to result from a complex interplay of facilitatory and inhibitory actions at different brain sites in the anxiety/fear circuits. To better understand the effect of mGlu7 and mGlu8 receptors on extinction of contextual fear and their critical sites of action in the fear networks, we focused on the amygdala. Direct injection into the basolateral complex of the amygdala of the mGlu7 receptor agonist AMN082 facilitated extinction, whereas the mGlu8 receptor agonist (S)-3,4-DCPG sustained freezing during the extinction acquisition trial. We also determined at the ultrastructural level the synaptic distribution of these receptors in the basal nucleus (BA) and intercalated cell clusters (ITCs) of the amygdala. Both areas are thought to exert key roles in fear extinction. We demonstrate that mGlu7 and mGlu8 receptors are located in different presynaptic terminals forming both asymmetric and symmetric synapses, and that they preferentially target neurons expressing mGlu1α receptors mostly located around ITCs. In addition we show that mGlu7 and mGlu8 receptors were segregated to different inputs to a significant extent. In particular, mGlu7a receptors were primarily onto glutamatergic afferents arising from the BA or midline thalamic nuclei, but not the medial prefrontal cortex (mPFC), as revealed by combined anterograde tracing and pre-embedding electron microscopy. On the other hand, mGlu8a showed a more restricted distribution in the BA and appeared absent from thalamic, mPFC and intrinsic inputs. This segregation of mGlu7 and mGlu8 receptors in different neuronal pathways of the fear circuit might explain the distinct effects on fear extinction training observed with mGlu7 and mGlu8 receptor agonists. This article is part of a Special Issue entitled 'Metabotropic Glutamate Receptors'.
Collapse
Affiliation(s)
- Alice Dobi
- Division of Cerebral Structure, National Institute for Physiological Sciences, Myodaiji, Okazaki 444-8787, Japan
| | | | | | | | | | | | | |
Collapse
|
137
|
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
|
138
|
Fazio F, Lionetto L, Molinaro G, Bertrand HO, Acher F, Ngomba RT, Notartomaso S, Curini M, Rosati O, Scarselli P, Di Marco R, Battaglia G, Bruno V, Simmaco M, Pin JP, Nicoletti F, Goudet C. Cinnabarinic acid, an endogenous metabolite of the kynurenine pathway, activates type 4 metabotropic glutamate receptors. Mol Pharmacol 2012; 81:643-56. [PMID: 22311707 DOI: 10.1124/mol.111.074765] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cinnabarinic acid is an endogenous metabolite of the kynurenine pathway that meets the structural requirements to interact with glutamate receptors. We found that cinnabarinic acid acts as a partial agonist of type 4 metabotropic glutamate (mGlu4) receptors, with no activity at other mGlu receptor subtypes. We also tested the activity of cinnabarinic acid on native mGlu4 receptors by examining 1) the inhibition of cAMP formation in cultured cerebellar granule cells; 2) protection against excitotoxic neuronal death in mixed cultures of cortical cells; and 3) protection against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine toxicity in mice after local infusion into the external globus pallidus. In all these models, cinnabarinic acid behaved similarly to conventional mGlu4 receptor agonists, and, at least in cultured neurons, the action of low concentrations of cinnabarinic acid was largely attenuated by genetic deletion of mGlu4 receptors. However, high concentrations of cinnabarinic acid were still active in the absence of mGlu4 receptors, suggesting that the compound may have off-target effects. Mutagenesis and molecular modeling experiments showed that cinnabarinic acid acts as an orthosteric agonist interacting with residues of the glutamate binding pocket of mGlu4. Accordingly, cinnabarinic acid did not activate truncated mGlu4 receptors lacking the N-terminal Venus-flytrap domain, as opposed to the mGlu4 receptor enhancer, N-phenyl-7-(hydroxyimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC). Finally, we could detect endogenous cinnabarinic acid in brain tissue and peripheral organs by high-performance liquid chromatography-tandem mass spectrometry analysis. Levels increased substantially during inflammation induced by lipopolysaccharide. We conclude that cinnabarinic acid is a novel endogenous orthosteric agonist of mGlu4 receptors endowed with neuroprotective activity.
Collapse
Affiliation(s)
- F Fazio
- Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Hinoi E, Yoneda Y. [Glutamate signaling in non-neuronal tissues]. Nihon Yakurigaku Zasshi 2012; 139:165-9. [PMID: 22498681 DOI: 10.1254/fpj.139.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
140
|
Koh DS, Cho JH, Chen L. Paracrine interactions within islets of Langerhans. J Mol Neurosci 2012; 48:429-40. [PMID: 22528452 DOI: 10.1007/s12031-012-9752-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 03/12/2012] [Indexed: 01/05/2023]
Abstract
Glucose supply fluctuates between meal and fasting periods and its consumption by the body varies greatly depending on bodily metabolism. Pancreatic islets of Langerhans secrete various endocrine hormones including insulin and glucagon to keep blood glucose level relatively constant. Additionally, islet hormones regulate activity of neighboring cells as local autocrine or paracrine modulators. Moreover, islet cells release neurotransmitters such as glutamate and γ-aminobutyric acid (GABA) to gain more precise regulation of hormones release kinetics. Excitatory glutamate is co-released with glucagon from α-cells and activates glutamate receptors in the neighboring cells. GABA released from β-cells was shown to inhibit α-cells but to activate β-cells by acting GABA(A) receptors. This review summarizes the recent progress in understanding the paracrine/autocrine interactions in islets.
Collapse
Affiliation(s)
- Duk-Su Koh
- University of Washington, Seattle, WA, USA.
| | | | | |
Collapse
|
141
|
Herman EJ, Bubser M, Conn PJ, Jones CK. Metabotropic glutamate receptors for new treatments in schizophrenia. Handb Exp Pharmacol 2012:297-365. [PMID: 23027420 DOI: 10.1007/978-3-642-25758-2_11] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Metabotropic glutamate receptors (mGluRs) represent exciting targets for the development of novel therapeutic agents for schizophrenia. Recent studies indicate that selective activation of specific mGluR subtypes may provide potential benefits for not only the positive symptoms, but also the negative symptoms and cognitive impairments observed in individuals with schizophrenia. Although optimization of traditional orthosteric agonists may still offer a feasible approach for the activation of mGluRs, important progress has been made in the discovery of novel subtype-selective allosteric ligands, including positive allosteric modulators (PAMs) of mGluR2 and mGluR5. These allosteric mGluR ligands have improved properties for clinical development and have served as key preclinical tools for a more in-depth understanding of the potential roles of these different mGluR subtypes for the treatment of schizophrenia.
Collapse
Affiliation(s)
- E J Herman
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
142
|
High glucose stimulates glutamate uptakes in pancreatic β-cells. Lab Anim Res 2011; 27:327-31. [PMID: 22232641 PMCID: PMC3251763 DOI: 10.5625/lar.2011.27.4.327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Revised: 12/02/2011] [Accepted: 12/12/2011] [Indexed: 02/07/2023] Open
Abstract
Pancreatic β-cells are major cells responsible for glucose metabolism in the body. Hyperglycemia is known to be a primary factor in the induction of diabetes mellitus. Glutamate is also an excitatory neurotransmitter in diverse organs. Oxidative stress also plays a pivotal role in the development of diabetes mellitus. However, the effect of hyperglycemia in glutamate uptake in the pancreas is not clear. Furthermore, the relationship between high glucose-induced glutamate uptake and oxidative stress has not been investigated. Therefore, this study was conducted to investigate the effect of high glucose on glutamate uptake in pancreatic β-cells. In the present study, 25 mM glucose stimulated the glutamate uptake in HIT-15 cells of hamster pancreatic β-cells. The treatment of 25 mM glucose and 1 mM glutamate also decreased the cell viability in HIT-15 cells. In addition, the treatment of 25 mM glucose induced an increase of lipid peroxide formation. High glucose-induced increase of LPO formation was prevented by the treatment of antioxidants such as N-acetyl-L-cysteine and quercetin. Furthermore, high glucose-induced stimulation of glutamate uptake and decrease of cell viability were also blocked by the treatment of N-acetyl-L-cysteine and quercetin. In conclusion, high glucose stimulated glutamate uptake via oxidative stress in pancreatic β-cells.
Collapse
|
143
|
|
144
|
Friedman M, Levin CE. Nutritional and medicinal aspects of D-amino acids. Amino Acids 2011; 42:1553-82. [PMID: 21519915 DOI: 10.1007/s00726-011-0915-1] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 04/06/2011] [Indexed: 02/07/2023]
Abstract
This paper reviews and interprets a method for determining the nutritional value of D-amino acids, D-peptides, and amino acid derivatives using a growth assay in mice fed a synthetic all-amino acid diet. A large number of experiments were carried out in which a molar equivalent of the test compound replaced a nutritionally essential amino acid such as L-lysine (L-Lys), L-methionine (L-Met), L-phenylalanine (L-Phe), and L-tryptophan (L-Trp) as well as the semi-essential amino acids L-cysteine (L-Cys) and L-tyrosine (L-Tyr). The results show wide-ranging variations in the biological utilization of test substances. The method is generally applicable to the determination of the biological utilization and safety of any amino acid derivative as a potential nutritional source of the corresponding L-amino acid. Because the organism is forced to use the D-amino acid or amino acid derivative as the sole source of the essential or semi-essential amino acid being replaced, and because a free amino acid diet allows better control of composition, the use of all-amino-acid diets for such determinations may be preferable to protein-based diets. Also covered are brief summaries of the widely scattered literature on dietary and pharmacological aspects of 27 individual D-amino acids, D-peptides, and isomeric amino acid derivatives and suggested research needs in each of these areas. The described results provide a valuable record and resource for further progress on the multifaceted aspects of D-amino acids in food and biological samples.
Collapse
Affiliation(s)
- Mendel Friedman
- Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| | | |
Collapse
|
145
|
Sheffler DJ, Gregory KJ, Rook JM, Conn PJ. Allosteric modulation of metabotropic glutamate receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2011; 62:37-77. [PMID: 21907906 DOI: 10.1016/b978-0-12-385952-5.00010-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The development of receptor subtype-selective ligands by targeting allosteric sites of G protein-coupled receptors (GPCRs) has proven highly successful in recent years. One GPCR family that has greatly benefited from this approach is the metabotropic glutamate receptors (mGlus). These family C GPCRs participate in the neuromodulatory actions of glutamate throughout the CNS, where they play a number of key roles in regulating synaptic transmission and neuronal excitability. A large number of mGlu subtype-selective allosteric modulators have been identified, the majority of which are thought to bind within the transmembrane regions of the receptor. These modulators can either enhance or inhibit mGlu functional responses and, together with mGlu knockout mice, have furthered the establishment of the physiologic roles of many mGlu subtypes. Numerous pharmacological and receptor mutagenesis studies have been aimed at providing a greater mechanistic understanding of the interaction of mGlu allosteric modulators with the receptor, which have revealed evidence for common allosteric binding sites across multiple mGlu subtypes and the presence for multiple allosteric sites within a single mGlu subtype. Recent data have also revealed that mGlu allosteric modulators can display functional selectivity toward particular signal transduction cascades downstream of an individual mGlu subtype. Studies continue to validate the therapeutic utility of mGlu allosteric modulators as a potential therapeutic approach for a number of disorders including anxiety, schizophrenia, Parkinson's disease, and Fragile X syndrome.
Collapse
Affiliation(s)
- Douglas J Sheffler
- Department of Pharmacology, Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | |
Collapse
|
146
|
Nicoletti F, Bockaert J, Collingridge GL, Conn PJ, Ferraguti F, Schoepp DD, Wroblewski JT, Pin JP. Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology 2010; 60:1017-41. [PMID: 21036182 DOI: 10.1016/j.neuropharm.2010.10.022] [Citation(s) in RCA: 494] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 10/15/2010] [Accepted: 10/21/2010] [Indexed: 12/24/2022]
Abstract
Metabotropic glutamate (mGlu) receptors were discovered in the mid 1980s and originally described as glutamate receptors coupled to polyphosphoinositide hydrolysis. Almost 6500 articles have been published since then, and subtype-selective mGlu receptor ligands are now under clinical development for the treatment of a variety of disorders such as Fragile-X syndrome, schizophrenia, Parkinson's disease and L-DOPA-induced dyskinesias, generalized anxiety disorder, chronic pain, and gastroesophageal reflux disorder. Prof. Erminio Costa was linked to the early times of the mGlu receptor history, when a few research groups challenged the general belief that glutamate could only activate ionotropic receptors and all metabolic responses to glutamate were secondary to calcium entry. This review moves from those nostalgic times to the most recent advances in the physiology and pharmacology of mGlu receptors, and highlights the role of individual mGlu receptor subtypes in the pathophysiology of human disorders. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.
Collapse
Affiliation(s)
- F Nicoletti
- Department of Physiology and Pharmacology, University of Rome, Sapienza, Piazzale Aldo Moro 5, 00185 Rome, Italy.
| | | | | | | | | | | | | | | |
Collapse
|