1
|
Ibrahim KS, Abd-Elrahman KS, El Mestikawy S, Ferguson SSG. Targeting Vesicular Glutamate Transporter Machinery: Implications on Metabotropic Glutamate Receptor 5 Signaling and Behavior. Mol Pharmacol 2020; 98:314-327. [PMID: 32873747 DOI: 10.1124/molpharm.120.000089] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/10/2020] [Indexed: 11/22/2022] Open
Abstract
Cross talk between both pre- and postsynaptic components of glutamatergic neurotransmission plays a crucial role in orchestrating a multitude of brain functions, including synaptic plasticity and motor planning. Metabotropic glutamate receptor (mGluR) 5 exhibits promising therapeutic potential for many neurodevelopmental and neurodegenerative disorders as a consequence of its modulatory control over diverse neuronal networks required for memory, motor coordination, neuronal survival, and differentiation. Given these crucial roles, mGluR5 signaling is under the tight control of glutamate release machinery mediated through vesicular glutamate transporters (VGLUTs) that ultimately dictate glutamatergic output. A particular VGLUT isoform, VGLUT3, exhibits an overlapping, but unique, distribution with mGluR5, and the dynamic cross talk between mGluR5 and VGLUT3 is key for the function of specific neuronal networks involved in motor coordination, emotions, and cognition. Thus, aberrant signaling of the VGLUT3-mGluR5 axis is linked to various pathologies including, but not limited to, Parkinson disease, anxiety disorders, and drug addiction. We argue that a comprehensive profiling of how coordinated VGLUT3-mGluR5 signaling influences overall glutamatergic neurotransmission is warranted. SIGNIFICANCE STATEMENT: Vesicular glutamate receptor (VGLUT) 3 machinery orchestrates glutamate release, and its distribution overlaps with metabotropic glutamate receptor (mGluR) 5 in regional brain circuitries, including striatum, hippocampus, and raphe nucleus. Therefore, VGLUT3-mGluR5 cross talk can significantly influence both physiologic and pathophysiologic glutamatergic neurotransmission. Pathological signaling of the VGLUT3-mGluR5 axis is linked to Parkinson disease, anxiety disorders, and drug addiction. However, it is also predicted to contribute to other motor and cognitive disorders.
Collapse
Affiliation(s)
- Karim S Ibrahim
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Salah El Mestikawy
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Institute (K.S.I., K.S.A.-E., S.S.G.F.) and Department of Cellular and Molecular Medicine (K.S.I., K.S.A.-E., S.S.G.F.), University of Ottawa, Ottawa, Ontario, Canada; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt (K.S.I., K.S.A.-E.); Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris, France (S.E.M.); and Department of Psychiatry, Douglas Hospital Research Center, McGill University, Verdun, Quebec, Canada (S.E.M.)
| |
Collapse
|
2
|
Abstract
Glutamate is the most abundant excitatory neurotransmitter, present at the bulk of cortical synapses, and participating in many physiologic and pathologic processes ranging from learning and memory to stroke. The tripeptide, glutathione, is one-third glutamate and present at up to low millimolar intracellular concentrations in brain, mediating antioxidant defenses and drug detoxification. Because of the substantial amounts of brain glutathione and its rapid turnover under homeostatic control, we hypothesized that glutathione is a relevant reservoir of glutamate and could influence synaptic excitability. We find that drugs that inhibit generation of glutamate by the glutathione cycle elicit decreases in cytosolic glutamate and decreased miniature excitatory postsynaptic potential (mEPSC) frequency. In contrast, pharmacologically decreasing the biosynthesis of glutathione leads to increases in cytosolic glutamate and enhanced mEPSC frequency. The glutathione cycle can compensate for decreased excitatory neurotransmission when the glutamate-glutamine shuttle is inhibited. Glutathione may be a physiologic reservoir of glutamate neurotransmitter.
Collapse
|
3
|
Zhang FX, Ge SN, Dong YL, Shi J, Feng YP, Li Y, Li YQ, Li JL. Vesicular glutamate transporter isoforms: The essential players in the somatosensory systems. Prog Neurobiol 2018; 171:72-89. [PMID: 30273635 DOI: 10.1016/j.pneurobio.2018.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 08/28/2018] [Accepted: 09/23/2018] [Indexed: 02/08/2023]
Abstract
In nervous system, glutamate transmission is crucial for centripetal conveyance and cortical perception of sensory signals of different modalities, which necessitates vesicular glutamate transporters 1-3 (VGLUT 1-3), the three homologous membrane-bound protein isoforms, to load glutamate into the presysnaptic vesicles. These VGLUTs, especially VGLUT1 and VGLUT2, selectively label and define functionally distinct neuronal subpopulations at each relay level of the neural hierarchies comprising spinal and trigeminal sensory systems. In this review, by scrutinizing each structure of the organism's fundamental hierarchies including dorsal root/trigeminal ganglia, spinal dorsal horn/trigeminal sensory nuclear complex, somatosensory thalamic nuclei and primary somatosensory cortex, we summarize and characterize in detail within each relay the neuronal clusters expressing distinct VGLUT protein/transcript isoforms, with respect to their regional distribution features (complementary distribution in some structures), axonal terminations/peripheral innervations and physiological functions. Equally important, the distribution pattern and characteristics of VGLUT1/VGLUT2 axon terminals within these structures are also epitomized. Finally, the correlation of a particular VGLUT isoform and its physiological role, disclosed thus far largely via studying the peripheral receptors, is generalized by referring to reports on global and conditioned VGLUT-knockout mice. Also, researches on VGLUTs relating to future direction are tentatively proposed, such as unveiling the elusive differences between distinct VGLUTs in mechanism and/or pharmacokinetics at ionic/molecular level, and developing VGLUT-based pain killers.
Collapse
Affiliation(s)
- Fu-Xing Zhang
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Shun-Nan Ge
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yu-Lin Dong
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Juan Shi
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yu-Peng Feng
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China
| | - Yang Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, PR China
| | - Yun-Qing Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China; Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, PR China.
| | - Jin-Lian Li
- Department of Anatomy and K.K. Leung Brain Research Centre, School of Basic Medicine, The Fourth Military Medical University, Xi'an 710032, PR China.
| |
Collapse
|
4
|
Effective Mechanism for Synthesis of Neurotransmitter Glutamate and its Loading into Synaptic Vesicles. Neurochem Res 2016; 42:64-76. [DOI: 10.1007/s11064-016-2037-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 08/14/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023]
|
5
|
Pál I, Nyitrai G, Kardos J, Héja L. Neuronal and astroglial correlates underlying spatiotemporal intrinsic optical signal in the rat hippocampal slice. PLoS One 2013; 8:e57694. [PMID: 23469218 PMCID: PMC3585794 DOI: 10.1371/journal.pone.0057694] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Accepted: 01/28/2013] [Indexed: 11/24/2022] Open
Abstract
Widely used for mapping afferent activated brain areas in vivo, the label-free intrinsic optical signal (IOS) is mainly ascribed to blood volume changes subsequent to glial glutamate uptake. By contrast, IOS imaged in vitro is generally attributed to neuronal and glial cell swelling, however the relative contribution of different cell types and molecular players remained largely unknown. We characterized IOS to Schaffer collateral stimulation in the rat hippocampal slice using a 464-element photodiode-array device that enables IOS monitoring at 0.6 ms time-resolution in combination with simultaneous field potential recordings. We used brief half-maximal stimuli by applying a medium intensity 50 Volt-stimulus train within 50 ms (20 Hz). IOS was primarily observed in the str. pyramidale and proximal region of the str. radiatum of the hippocampus. It was eliminated by tetrodotoxin blockade of voltage-gated Na(+) channels and was significantly enhanced by suppressing inhibitory signaling with gamma-aminobutyric acid(A) receptor antagonist picrotoxin. We found that IOS was predominantly initiated by postsynaptic Glu receptor activation and progressed by the activation of astroglial Glu transporters and Mg(2+)-independent astroglial N-methyl-D-aspartate receptors. Under control conditions, role for neuronal K(+)/Cl(-) cotransporter KCC2, but not for glial Na(+)/K(+)/Cl(-) cotransporter NKCC1 was observed. Slight enhancement and inhibition of IOS through non-specific Cl(-) and volume-regulated anion channels, respectively, were also depicted. High-frequency IOS imaging, evoked by brief afferent stimulation in brain slices provide a new paradigm for studying mechanisms underlying IOS genesis. Major players disclosed this way imply that spatiotemporal IOS reflects glutamatergic neuronal activation and astroglial response, as observed within the hippocampus. Our model may help to better interpret in vivo IOS and support diagnosis in the future.
Collapse
Affiliation(s)
- Ildikó Pál
- Department of Functional Pharmacology, Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.
| | | | | | | |
Collapse
|
6
|
Callaerts-Vegh Z, Moechars D, Van Acker N, Daneels G, Goris I, Leo S, Naert A, Meert T, Balschun D, D'Hooge R. Haploinsufficiency of VGluT1 but not VGluT2 impairs extinction of spatial preference and response suppression. Behav Brain Res 2013; 245:13-21. [PMID: 23396167 DOI: 10.1016/j.bbr.2013.01.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Revised: 01/08/2013] [Accepted: 01/15/2013] [Indexed: 11/27/2022]
Abstract
The excitatory neurotransmitter l-glutamate is transported into synaptic vesicles by vesicular glutamate transporters (VGluTs) to transmit glutamatergic signals. Changes in their expression have been linked to various brain disorders including schizophrenia, Parkinson's, and Alzheimer's disease. Deleting either the VGluT1 or VGluT2 gene leads to profound developmental and neurological complications and early death, but mice heterozygous for VGluT1 or VGluT2 are viable and thrive. Acquisition, retention and extinction of conditioned visuospatial and emotional responses were compared between VGluT1(+/-) and VGluT2(+/-) mice, and their wildtype littermates, using different water maze procedures, appetitive scheduled conditioning, and conditioned fear protocols. The distinct brain expression profiles of the VGluT1 and -2 isoforms particularly in telencephalic structures, such as neocortex, hippocampus and striatum, are reflected in very specific behavioral changes. VGluT2(+/-) mice were unimpaired in spatial learning tasks and fear extinction. Conversely, VGluT1(+/-) mice displayed spatial extinction learning deficits and markedly impaired fear extinction. These data indicate that VGluT1, but not VGluT2, plays a role in the neural processes underlying inhibitory learning.
Collapse
|
7
|
Enhanced glutamate uptake into synaptic vesicles fueled by vesicle-generated ATP from phosphoenolpyruvate and ADP. Proposed role of a novel enzyme. Neurochem Res 2012; 37:2731-7. [PMID: 22915206 DOI: 10.1007/s11064-012-0864-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 07/28/2012] [Accepted: 07/31/2012] [Indexed: 10/28/2022]
Abstract
Glycolytic ATP synthesis by synaptic vesicles provides an efficient mechanism for fueling vesicular loading of the neurotransmitter glutamate. This is achieved in part by vesicle-bound pyruvate kinase. However, we have found that vesicular glutamate uptake, in the presence of the pyruvate kinase substrates ADP and phosphoenolpyruvate (PEP), substantially exceeds that caused by exogenous ATP. We propose that this much enhanced uptake is in part due to extra ATP produced via a mechanism involving a novel enzyme, PEP-dependent ADP synthase. We discuss implications for this enzyme in energy homeostasis and pathophysiology, as well as in efficient synaptic glutamate transmission.
Collapse
|
8
|
Takeda K, Ishida A, Takahashi K, Ueda T. Synaptic vesicles are capable of synthesizing the VGLUT substrate glutamate from α-ketoglutarate for vesicular loading. J Neurochem 2012; 121:184-96. [PMID: 22309504 DOI: 10.1111/j.1471-4159.2012.07684.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Synaptic vesicle loading of glutamate is a pivotal step in glutamate synaptic transmission. The molecular machinery responsible for this step is comprised of v-type proton-pump ATPase and a vesicular glutamate transporter. Recent evidence indicates that synaptic vesicles are endowed with glycolytic ATP-synthesizing enzymes, providing energy for immediate use by vesicle-bound proton-pump ATPase. In this study, we provide evidence that synaptic vesicles are also capable of synthesizing the vesicular glutamate transporter substrate glutamate, from α-ketoglutarate and l-aspartate (as the amino group donor); glutamate thus produced is taken up into vesicles. We also report a finding that α-ketoglutarate-derived glutamate uptake into synaptic vesicles and aspartate aminotransferase are inhibited by 2,3-pyrazinedicarboxylate. Evidence is given that this is a selective inhibitor for aspartate aminotransferase. These observations provide insight into understanding the nerve endings' mechanism for high efficiency in glutamate transmission. Finding this inhibitor may have implications for further experimentation on the role of α-ketoglutarate-derived glutamate in glutamate transmission.
Collapse
Affiliation(s)
- Kouji Takeda
- Molecular & Behavioral Neuroscience Institute, Medical School, The University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
9
|
Koga M, Serritella AV, Messmer MM, Hayashi-Takagi A, Hester LD, Snyder SH, Sawa A, Sedlak TW. Glutathione is a physiologic reservoir of neuronal glutamate. Biochem Biophys Res Commun 2011; 409:596-602. [PMID: 21539809 DOI: 10.1016/j.bbrc.2011.04.087] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 04/19/2011] [Indexed: 01/04/2023]
Abstract
Glutamate, the principal excitatory neurotransmitter of the brain, participates in a multitude of physiologic and pathologic processes, including learning and memory. Glutathione, a tripeptide composed of the amino acids glutamate, cysteine, and glycine, serves important cofactor roles in antioxidant defense and drug detoxification, but glutathione deficits occur in multiple neuropsychiatric disorders. Glutathione synthesis and metabolism are governed by a cycle of enzymes, the γ-glutamyl cycle, which can achieve intracellular glutathione concentrations of 1-10mM. Because of the considerable quantity of brain glutathione and its rapid turnover, we hypothesized that glutathione may serve as a reservoir of neural glutamate. We quantified glutamate in HT22 hippocampal neurons, PC12 cells and primary cortical neurons after treatment with molecular inhibitors targeting three different enzymes of the glutathione metabolic cycle. Inhibiting 5-oxoprolinase and γ-glutamyl transferase, enzymes that liberate glutamate from glutathione, leads to decreases in glutamate. In contrast, inhibition of γ-glutamyl cysteine ligase, which uses glutamate to synthesize glutathione, results in substantial glutamate accumulation. Increased glutamate levels following inhibition of glutathione synthesis temporally precede later effects upon oxidative stress.
Collapse
Affiliation(s)
- Minori Koga
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Meyer 4-137, 600 North Wolfe Street, Baltimore, MD 21287, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Baghbanzadeh A, Modirsaneie M, Emam G, Hajinezhad M. Microhandling of vesicular glutamate uptake modulate feeding in broilers. J Anim Physiol Anim Nutr (Berl) 2010; 94:74-7. [DOI: 10.1111/j.1439-0396.2008.00887.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
11
|
Magni DV, Furian AF, Oliveira MS, Souza MA, Lunardi F, Ferreira J, Mello CF, Royes LFF, Fighera MR. Kinetic characterization of
l‐
[
3
H]glutamate uptake inhibition and increase oxidative damage induced by glutaric acid in striatal synaptosomes of rats. Int J Dev Neurosci 2008; 27:65-72. [DOI: 10.1016/j.ijdevneu.2008.09.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Revised: 08/30/2008] [Accepted: 09/23/2008] [Indexed: 10/21/2022] Open
Affiliation(s)
- Danieli Valnes Magni
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Ana Flávia Furian
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaUniversidade Federal do Rio Grande do Sul90035‐003Porto AlegreRSBrazil
| | - Mauro Schneider Oliveira
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Programa de Pós‐graduação em Ciências Biológicas: BioquímicaUniversidade Federal do Rio Grande do Sul90035‐003Porto AlegreRSBrazil
| | - Mauren Assis Souza
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Centro de Educação Física e DesportosDepartamento de Métodos e Técnicas DesportivasUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Fabiane Lunardi
- Centro de Ciências Naturais e ExatasLaboratório de Neurotoxicidade, Departamento de QuímicaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Juliano Ferreira
- Centro de Ciências Naturais e ExatasLaboratório de Neurotoxicidade, Departamento de QuímicaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Carlos Fernando Mello
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Luiz Fernando Freire Royes
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Centro de Ciências Naturais e ExatasLaboratório de Neurotoxicidade, Departamento de QuímicaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Centro de Educação Física e DesportosDepartamento de Métodos e Técnicas DesportivasUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
| | - Michele Rechia Fighera
- Centro de Ciências da SaúdeLaboratório de Psicofarmacologia e Neurotoxicidade, Departamento de FisiologiaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Centro de Ciências da SaúdeDepartamento de PediatriaUniversidade Federal de Santa Maria97105‐900Santa MariaRSBrazil
- Universidade Luterana do BrasilCampus Santa MariaSanta MariaRSBrazil
| |
Collapse
|
12
|
Synaptic vesicle-bound pyruvate kinase can support vesicular glutamate uptake. Neurochem Res 2008; 34:807-18. [PMID: 18751889 DOI: 10.1007/s11064-008-9833-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Accepted: 08/07/2008] [Indexed: 10/21/2022]
Abstract
Glucose metabolism is essential for normal brain function and plays a vital role in synaptic transmission. Recent evidence suggests that ATP synthesized locally by glycolysis, particularly via glyceraldehyde 3-phosphate dehydrogenase/3-phosphoglycerate kinase, is critical for synaptic transmission. We present evidence that ATP generated by synaptic vesicle-associated pyruvate kinase is harnessed to transport glutamate into synaptic vesicles. Isolated synaptic vesicles incorporated [(3)H]glutamate in the presence of phosphoenolpyruvate (PEP) and ADP. Pyruvate kinase activators and inhibitors stimulated and reduced PEP/ADP-dependent glutamate uptake, respectively. Membrane potential was also formed in the presence of pyruvate kinase activators. "ATP-trapping" experiments using hexokinase and glucose suggest that ATP produced by vesicle-associated pyruvate kinase is more readily used than exogenously added ATP. Other neurotransmitters such as GABA, dopamine, and serotonin were also taken up into crude synaptic vesicles in a PEP/ADP-dependent manner. The possibility that ATP locally generated by glycolysis supports vesicular accumulation of neurotransmitters is discussed.
Collapse
|
13
|
Li T, Bai L, Li J, Igarashi S, Ghishan FK. Sp1 is required for glucose-induced transcriptional regulation of mouse vesicular glutamate transporter 2 gene. Gastroenterology 2008; 134:1994-2003. [PMID: 18440316 PMCID: PMC2747381 DOI: 10.1053/j.gastro.2008.02.076] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 02/13/2008] [Accepted: 02/26/2008] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Vesicular glutamate transporter (VGLUT) has been reported to be involved in glucose-induced insulin secretion. It has been shown that glucose stimulates the expression of VGLUT isoform 2 (VGLUT2) in beta cells via transcriptional mechanism. In this study, we identified the mouse VGLUT2 (mVGLUT2) promoter and characterized the transcriptional mechanism of glucose-stimulated mVGLUT2 expression in beta-cells. METHODS A promoter region of mVGLUT2 was cloned by genomic polymerase chain reaction. The mechanism of Sp1 in glucose-induced transactivation of mVGLUT2 was investigated by luciferase assay, electrophoretic mobility shift assay, chromatin immunoprecipitation assay, and Western blot analysis. RESULTS A promoter containing 2133 base pairs of upstream sequence of the 5'-flanking region of mVGLUT2 complementary DNA was cloned. Transient transfection of various 5'-end deletion constructs of the mVGLUT2 promoter/luciferase reporter indicated that the region between -96 to +68 base pair contains the basal promoter for mVGLUT2. Mutational analysis and electromobility shift assay showed an important role for the transcription factor Sp1 in both basal and glucose-induced mVGLUT2 transcription. The interaction between Sp1 and mVGLUT2 was confirmed by chromatin immunoprecipitation assays. Glucose stimulates the phosphorylation of Sp1 via mitogen-activated protein kinase P38 and P44/42. This leads to increased binding activity of Sp1 to the mVGLUT2 promoter and results in activation of the gene. CONCLUSIONS We cloned the mouse VGLUT2 promoter and showed a novel molecular mechanism of glucose-induced mVGLUT2 transcription.
Collapse
Affiliation(s)
- Tao Li
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724
| | - Liqun Bai
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724,Department of Medicine, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724
| | - Jing Li
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724
| | - Suzu Igarashi
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724
| | - Fayez K. Ghishan
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona Health Science Center, Tucson, Arizona 85724,Corresponding author: Fayez K. Ghishan, M.D., Professor and Head, Department of Pediatrics, Steele Children's Research Center, University of Arizona Health Sciences Center, 1501 N. Campbell Ave., Tucson, AZ 85724, E-mail:
| |
Collapse
|
14
|
Winter HC, Ueda T. The glutamate uptake system in presynaptic vesicles: further characterization of structural requirements for inhibitors and substrates. Neurochem Res 2007; 33:223-31. [PMID: 17940888 DOI: 10.1007/s11064-007-9493-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 08/28/2007] [Indexed: 12/01/2022]
Abstract
Noncyclic fluorine-substituted and cyclic analogs of glutamic acid were tested for their ability to inhibit glutamate uptake in isolated bovine presynaptic vesicles, in order to assess the specific structural requirements of the glutamate translocation system in the vesicle membrane. Cyclic analogs that permitted close interaction between the positive and negative charges of the glutamate molecule were effective inhibitors; maximum inhibitory potency was observed with L-trans-1-aminocyclopentane-1,3-dicarboxylic acid (L-t-ACPD), while D-t-ACPD was less active. Analogs with a larger or smaller ring (as in trans-1-aminocyclohexane-1,3-dicarboxylic acid or trans-1-aminocyclobutane-1,3-dicarboxylic acid) were also inhibitory, but somewhat less so. trans-ACPD was also taken up by the vesicles with a time course and ATP dependence similar to uptake of glutamate, and this uptake was inhibited by glutamate. The K(m) value for t-ACPD uptake was similar to its K(i) for inhibition of glutamate uptake, while its rate of uptake was lower than that of glutamate. Fluorine-substituted noncyclic analogs with substitutions at the 4-carbon were less effective than glutamic acid itself, although 4,4-difluoroglutamic acid was equal in activity to the unsubstituted compound. Inhibition by these derivatives appeared to be competitive in nature, and they probably were also transported by the vesicle uptake system.
Collapse
Affiliation(s)
- Harry C Winter
- Molecular and Behavioral Neuroscience Institute, The University of Michigan Medical School, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA
| | | |
Collapse
|
15
|
Almqvist J, Huang Y, Laaksonen A, Wang DN, Hovmöller S. Docking and homology modeling explain inhibition of the human vesicular glutamate transporters. Protein Sci 2007; 16:1819-29. [PMID: 17660252 PMCID: PMC2206968 DOI: 10.1110/ps.072944707] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
As membrane transporter proteins, VGLUT1-3 mediate the uptake of glutamate into synaptic vesicles at presynaptic nerve terminals of excitatory neural cells. This function is crucial for exocytosis and the role of glutamate as the major excitatory neurotransmitter in the central nervous system. The three transporters, sharing 76% amino acid sequence identity in humans, are highly homologous but differ in regional expression in the brain. Although little is known regarding their three-dimensional structures, hydropathy analysis on these proteins predicts 12 transmembrane segments connected by loops, a topology similar to other members in the major facilitator superfamily, where VGLUT1-3 have been phylogenetically classified. In this work, we present a three-dimensional model for the human VGLUT1 protein based on its distant bacterial homolog in the same superfamily, the glycerol-3-phosphate transporter from Escherichia coli. This structural model, stable during molecular dynamics simulations in phospholipid bilayers solvated by water, reveals amino acid residues that face its pore and are likely to affect substrate translocation. Docking of VGLUT1 substrates to this pore localizes two different binding sites, to which inhibitors also bind with an overall trend in binding affinity that is in agreement with previously published experimental data.
Collapse
Affiliation(s)
- Jonas Almqvist
- Division of Structural Chemistry, Arrhenius Laboratory, Stockholm University, S-10691 Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
16
|
Abstract
Mammalian central synapses commonly specialize in one fast neurotransmitter, matching the content of their presynaptic vesicles with the appropriate receptors in their postsynaptic membrane. Here, I show that hippocampal cultures contain autaptic glutamatergic synapses that contravene this rule: in addition to postsynaptic glutamate receptors, they also express clusters of functional postsynaptic GABA(A) receptors yet lack presynaptic GABA. Hence, these synapses are presynaptically silent with respect to GABA. They can be unsilenced by loading GABA into presynaptic vesicles by endocytosis, after which a postload IPSC appears. This IPSC is similar to native IPSCs recorded from GABAergic interneurons in the same cultures. Thus, these "mistargeted" GABA(A) receptors, which apparently lack a signal that confers synaptic specificity, function almost normally. After GABA loading, glutamatergic miniature postsynaptic currents acquire a slow tail that is mediated by GABA(A) receptors, showing that synaptic vesicles can accommodate both the usual concentration of native glutamate and a saturating concentration of loaded GABA. After brief Ca(2+)-dependent exocytosis, endocytosis of GABA can proceed in low-Ca(2+) external solution. The amplitude of the postload IPSC declines exponentially with repetitive stimulation as the endocytosed GABA passes through the presynaptic vesicle cycle and is depleted. Hence, by using GABA as an exogenous but physiological tracer, the properties of these presynaptically silent synapses can provide novel insights into the content and cycling of vesicles in presynaptic terminals.
Collapse
|
17
|
Nickerson Poulin A, Guerci A, El Mestikawy S, Semba K. Vesicular glutamate transporter 3 immunoreactivity is present in cholinergic basal forebrain neurons projecting to the basolateral amygdala in rat. J Comp Neurol 2006; 498:690-711. [PMID: 16917846 DOI: 10.1002/cne.21081] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The basal forebrain (BF) plays a role in behavioral and cortical arousal, attention, learning, and memory. It has been suggested that cholinergic BF neurons co-release glutamate, and some cholinergic BF neurons have been reported to contain vesicular glutamate transporter 3 (VGLUT3). We examined the distribution and projections of BF cholinergic neurons containing VGLUT3, by using dual-label immunofluorescence for choline acetyltransferase (ChAT) and VGLUT3, in situ hybridization, and retrograde tracing. Neurons immunoreactive (+) or containing mRNAs for both ChAT and VGLUT3 were mainly localized to the ventral pallidum and more caudal BF regions; the co-immunoreactive neurons represented 31% of cholinergic neurons in the ventral pallidum and 5-9% more caudally. Examination of cholinergic axon terminals in known target areas of BF projections indicated that the basolateral amygdaloid nucleus contained numerous terminals co-immunoreactive for ChAT and VGLUT3, whereas sampled areas of the olfactory bulb, neocortex, hippocampus, reticular thalamic nucleus, and interpeduncular nucleus were devoid of double-labeled terminals. The basolateral amygdala is innervated by cholinergic BF neurons lacking low-affinity p75 nerve growth factor receptors; many ChAT+VGLUT3+ BF neurons were immunonegative to this receptor. Twenty-five to 79% of ChAT+VGLUT3+ neurons in different BF regions were retrogradely labeled from the basolateral amygdala, up to 52% (ventral pallidum) of the retrogradely labeled ChAT+ neurons were VGLUT3+, and the largest number of amygdala-projecting ChAT+VGluT3+ neurons was found in the ventral pallidum. These findings indicate that BF cholinergic neurons containing VGLUT3 project to the basolateral amygdala and suggest that these neurons might have the capacity to release both acetylcholine and glutamate.
Collapse
Affiliation(s)
- Amanda Nickerson Poulin
- Department of Anatomy and Neurobiology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 1X5, Canada
| | | | | | | |
Collapse
|
18
|
Aronoff R, Mellem JE, Maricq AV, Sprengel R, Seeburg PH. Neuronal toxicity in Caenorhabditis elegans from an editing site mutant in glutamate receptor channels. J Neurosci 2005; 24:8135-40. [PMID: 15371514 PMCID: PMC6729790 DOI: 10.1523/jneurosci.2587-04.2004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ionotropic glutamate receptors (iGluRs) in Caenorhabditis elegans are predicted to have high permeability for Ca2+ because of glutamine (Q) residues in the pore loop. This contrasts to the low Ca2+ permeability of similar iGluRs in principal neurons of mammals, because of an edited arginine (R) at the critical pore position in at least one channel subunit. Here, we introduced the R residue into the pore loop of a glutamate receptor subunit, GLR-2, in C. elegans. GLR-2(R) participated in channel formation, as revealed by decreased rectification of kainate-evoked currents in electrophysiological recordings when GLR-2(R) and the wild-type GLR-2(Q) were coexpressed in worms. Notably, the transgenic worms exhibited, at low penetrance, strong phenotypic impairments including uncoordination, neuronal degeneration, developmental arrest, and lethality. Penetrance of adverse phenotypes could be enhanced by transgenic expression of an optimal GLR-2(Q)/(R) ratio, implicating channel activity as the cause. In direct support, a mutation in eat-4, which prevents glutamatergic transmission, suppressed adverse phenotypes. Suppression was also achieved by mutation in calreticulin, which is necessary for maintainance of intracellular Ca2+ stores in the endoplasmic reticulum. Thus, synaptically activated GLR-2(R)-containing iGluR channels appear to trigger inappropriate, neurotoxic Ca2+ release from intracellular stores.
Collapse
Affiliation(s)
- Rachel Aronoff
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
19
|
Abstract
Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system (CNS). Packaging and storage of glutamate into glutamatergic neuronal vesicles require ATP-dependent vesicular glutamate uptake systems, which utilize the electrochemical proton gradient as a driving force. Three vesicular glutamate transporters (VGLUT1-3) have been recently identified from neuronal tissue where they play a key role to maintain the vesicular glutamate level. Recently, it has been demonstrated that glutamate signaling is also functional in peripheral neuronal and non-neuronal tissues, and occurs in sites of pituitary, adrenal, pineal glands, bone, GI tract, pancreas, skin, and testis. The glutamate receptors and VGLUTs in digestive system have been found in both neuronal and endocrinal cells. The glutamate signaling in the digestive system may have significant relevance to diabetes and GI tract motility disorders. This review will focus on the most recent update of molecular physiology of digestive VGLUTs.
Collapse
Affiliation(s)
- Tao Li
- Departments of Pediatrics, Room 3325, Steele Memorial Children's Research Center, University of Arizona Health Sciences Center, Tucson, AZ 85724, USA
| | | | | |
Collapse
|
20
|
Barbaro NM, Takahashi DK, Baraban SC. A potential role for astrocytes in mediating the antiepileptic actions of furosemide in vitro. Neuroscience 2005; 128:655-63. [PMID: 15381293 DOI: 10.1016/j.neuroscience.2004.07.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2004] [Indexed: 11/17/2022]
Abstract
Epileptic seizures are characterized by abnormal electrical discharge. In previous studies we established a powerful antiepileptic action for a commonly used diuretic (furosemide). However, it remains unclear precisely how furosemide terminates abnormal electrical discharges. To address this issue, we performed in vitro experiments to examine conditions where furosemide exerts antiepileptic activity and patch-clamp studies to analyze the effect of furosemide on neuronal membrane properties, synaptic function and inward potassium current. Furosemide was not found to alter synaptic field responses, excitatory postsynaptic currents or intrinsic membrane properties of principal hippocampal neurons. Our in vitro studies indicate that furosemide does not abolish spontaneous epileptiform bursting during co-application of Ba2+ or Cs+ ions (to block inwardly rectifying potassium channels). Our patch-clamp data indicate that furosemide enhances the function of astrocytic, but not neuronal, inward potassium channels and that this modulation may be required for its antiepileptic activity. Although a variety of antiepileptic drugs are already available, none of these compounds selectively target astrocytes while preserving synaptic/neuronal function. Thus, furosemide-mediated modulation of inward potassium current (on astrocytes) represents a new target for control of abnormal electrical discharge in the CNS.
Collapse
Affiliation(s)
- N M Barbaro
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, 513 Parnassus Avenue, Box 0520, San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
21
|
Bole DG, Ueda T. Inhibition of Vesicular Glutamate Uptake by Rose Bengal-Related Compounds: Structure–Activity Relationship. Neurochem Res 2005; 30:363-9. [PMID: 16018580 DOI: 10.1007/s11064-005-2610-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Synaptic vesicular accumulation of glutamate is a vital initial step in glutamate transmission. We have previously shown that Rose Bengal, a polyhalogenated fluorescein analog, is a potent inhibitor of glutamate uptake into synaptic vesicles. Here, we report the structural features of Rose Bengal required for this inhibition. Various Rose Bengal-related compounds, with systematic structural variations, were tested. Results indicate that the four iodo groups and the phenyl group attached to the xanthene moiety are critical for potent inhibitory activity. Replacement of these groups with two iodo groups and an alkyl group, respectively, results in substantial reduction in potency. Of further interest in creating high potency is the critical nature of the oxygen atom which links the two benzene rings of xanthene. Thus, the phenyl group and multiple iodo groups, as well as the bridging oxygen of xanthene, are crucial elements of Rose Bengal required for its potent inhibitory action.
Collapse
Affiliation(s)
- David G Bole
- Mental Health Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109-0669, USA
| | | |
Collapse
|
22
|
Porciúncula LO, Emanuelli T, Tavares RG, Schwarzbold C, Frizzo MES, Souza DO, Wajner M. Glutaric acid stimulates glutamate binding and astrocytic uptake and inhibits vesicular glutamate uptake in forebrain from young rats. Neurochem Int 2004; 45:1075-86. [PMID: 15337307 DOI: 10.1016/j.neuint.2004.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2003] [Revised: 04/23/2004] [Accepted: 05/03/2004] [Indexed: 11/16/2022]
Abstract
Glutaric acidemia type I (GA I) is an inherited neurometabolic disorder caused by glutaryl-CoA dehydrogenase deficiency, which leads to accumulation in body fluids and in brain of predominantly glutaric acid (GA), and to a lesser extent of 3-hydroxyglutaric and glutaconic acids. Neurological presentation is common in patients with GA I. Although the mechanisms underlying brain damage in this disorder are not yet well established, there is growing evidence that excitotoxicity may play a central role in the neuropathogenesis of this disease. In the present study, preparations of synaptosomes, synaptic plasma membranes and synaptic vesicles, as well as cultured astrocytes from rat forebrain were exposed to various concentrations of GA for the determination of the basal and potassium-induced release of [(3)H]glutamate by synaptosomes, Na(+)-independent glutamate binding to synaptic membranes and vesicular glutamate uptake and Na(+)-dependent glutamate uptake into astrocytes, respectively. GA (1-100 nM) significantly stimulated [(3)H]glutamate binding to brain plasma membranes (40-70%) in the absence of extracellular Na(+) concentrations, reflecting glutamate binding to receptors. Furthermore, this stimulatory effect was totally abolished by the metabotropic glutamate ligands DHPG, DCG-IV and l-AP4, attenuated by the ionotropic non-NMDA glutamate receptor agonist AMPA and had no interference of the NMDA receptor antagonist MK-801. Moreover, [(3)H]glutamate uptake into synaptic vesicles was inhibited by approximately 50% by 10 and 100 nM GA and Na(+)-dependent [(3)H]glutamate uptake by astrocytes was significantly increased (up to 50%) in a dose-dependent manner (maximal stimulation at 100 microM GA). In contrast, synaptosomal glutamate release was not affected by the acid at concentrations as high as 1 mM. These results indicate that the inhibition of glutamate uptake into synaptic vesicles by low concentrations GA may result in elevated concentrations of the excitatory neurotransmitter in the cytosol and the stimulatory effect of this organic acid on glutamate binding may potentially cause excitotoxicity to neural cells. Finally, taken together these results and previous findings showing that GA markedly decreases synaptosomal glutamate uptake, it is possible that the stimulatory effect of GA on astrocyte glutamate uptake might indicate that astrocytes may protect neurons from excitotoxic damage caused by GA by increasing glutamate uptake and therefore reducing the concentration of this excitatory neurotransmitter in the synaptic cleft.
Collapse
Affiliation(s)
- Lisiane O Porciúncula
- Departamento de Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP 90035-003 Porto Alegre, RS, Brazil
| | | | | | | | | | | | | |
Collapse
|
23
|
Brouns I, Pintelon I, Van Genechten J, De Proost I, Timmermans JP, Adriaensen D. Vesicular glutamate transporter 2 is expressed in different nerve fibre populations that selectively contact pulmonary neuroepithelial bodies. Histochem Cell Biol 2003; 121:1-12. [PMID: 14677070 DOI: 10.1007/s00418-003-0609-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2003] [Indexed: 11/29/2022]
Abstract
Pulmonary neuroepithelial body (NEB) receptors in rats receive at least four different nerve fibre populations. In addition to a spinal sensory innervation that contacts NEBs at their basal side, extensive vagal nodose sensory terminals and separate nitrergic and cholinergic nerve endings protrude between NEB cells. In the present study, antibodies against the vesicular glutamate transporter 2 (VGLUT2), a transmembrane protein responsible for loading glutamate into synaptic vesicles, were used to investigate whether some of the nerve terminals contacting NEBs in rat lungs might use glutamate as a neurotransmitter. VGLUT2 immunoreactivity (IR) was detected in extensive intraepithelial arborising nerve terminals that appeared to contact most of the NEBs. Multiple immunostaining showed VGLUT2 IR in the vagal nodose and spinal sensory nerve terminals contacting NEBs, and in another, most likely sensory, intraepithelial nerve fibre population, the origin and further characteristics of which remain to be elucidated. At least part of the VGLUT2-immunoreactive nerve fibres that contact NEBs were shown to be myelinated. The expression of VGLUT2 indicates that glutamate is stored and released as a neurotransmitter in terminals of several pulmonary (sensory) nerve fibre populations that selectively relate to the complex NEB receptors. The present study strongly suggests an involvement of glutamatergic mechanisms in the peripheral transduction of sensory stimuli from the lungs, via the release of glutamate from nerve terminals, thereby modulating the activity of NEB receptor cells or the excitability of afferent nerves.
Collapse
Affiliation(s)
- Inge Brouns
- Laboratory of Cell Biology and Histology, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
24
|
Hayashi M, Morimoto R, Yamamoto A, Moriyama Y. Expression and localization of vesicular glutamate transporters in pancreatic islets, upper gastrointestinal tract, and testis. J Histochem Cytochem 2003; 51:1375-90. [PMID: 14500705 DOI: 10.1177/002215540305101014] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The wide-ranging expression of glutamate receptors in peripheral tissues suggests an unexpectedly wider role(s) of l-glutamate as an intercellular signaling molecule. However, the peripheral glutamatergic system is poorly understood, partly because the sites of l-glutamate signal appearance are less well characterized. Vesicular glutamate transporters (VGLUTs) are potential probes for the sites of vesicular storage and subsequent secretion of l-glutamate. In this study we raised specific polyclonal antibodies against two VGLUT isoforms, VGLUT1 and VGLUT2, and investigated their localization in peripheral tissues of rat. We detected the expression of either VGLUT1 or VGLUT2, or both, in pancreas, stomach, intestine, and testis. In pancreas, VGLUT1 and VGLUT2 are present in pancreatic polypeptide-containing secretory granules in F-cells in the islets of Langerhans. In stomach, VGLUT2 is abundant in the antrum and pylorus and is present in a subset of pancreatic polypeptide-containing cells. In intestine, VGLUT2 is abundant in the ileum and is co-localized with glucagon-like immunoreactive peptide and polypeptide YY (PYY). In testis, VGLUT2 is expressed and localized in the outer acrosomal membrane of spermatids, where KA1 and GluR5, kainate receptor subunits, are almost always localized. Taken together, these results strongly suggest the occurrence of a peripheral glutamatergic system in the gastroenteropancreatic system and testis.
Collapse
Affiliation(s)
- Mitsuko Hayashi
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | | | | | |
Collapse
|
25
|
Tong Q, Kirchgessner AL. Localization and function of metabotropic glutamate receptor 8 in the enteric nervous system. Am J Physiol Gastrointest Liver Physiol 2003; 285:G992-G1003. [PMID: 12829438 DOI: 10.1152/ajpgi.00118.2003] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The enteric nervous system (ENS) contains glutamatergic neurons, transporters, and functional ionotropic and groups I and II metabotropic glutamate receptors (mGluRs). The aim of this study was to determine whether the ENS contains functional group III mGluRs. RT-PCR demonstrated the expression of mGluR7 and mGluR8 mRNA in rat myenteric ganglia. Western blot analysis confirmed the presence of mGluR8 protein. Immunocytochemistry, in conjunction with confocal microscopy, demonstrated mGluR8 immunoreactivity in the ENS of several species, including humans. mGluR8 immunoreactivity was localized to the membrane of nerve cell bodies that received glutamatergic input. Significant receptor internalization of mGluR8 was observed on activation, and localization to membrane was observed on blocking with the mGluR III antagonist (RS)-cyclopropyl-4-phosphonophenylglycine (CPPG). mGluR8-positive myenteric neurons contained glutamate or nitric oxide synthase (NOS), a marker of inhibitory motorneurons. Enteric group III mGluRs are functional because mGluR8 agonists inhibited forskolin-induced accumulation of cAMP in isolated myenteric ganglia, and CPPG reduced this effect. In addition, an accelerating effect on guinea pig colonic motility was observed after the application of mGluR8 agonists. Increase in motility was specific, because CPPG inhibited it. Moreover, in the presence of hexamethonium or Nomega-nitro-l-arginine methyl ester, an inhibitor of NOS, responses caused by mGluR8 agonists were abolished. mGluR8 agonists also increased longitudinal muscle contractions. These findings suggest that mGluR8 agonists increase motility by inhibiting nitrergic relaxation and possibly by facilitating cholinergic contractions.
Collapse
Affiliation(s)
- Qingchun Tong
- Dept. of Physiology and Pharmacology, Box 29, State University of New York Downstate Medical Center, 450 Clarkson Ave., Brooklyn, NY 11203, USA.
| | | |
Collapse
|
26
|
Rosin DL, Weston MC, Sevigny CP, Stornetta RL, Guyenet PG. Hypothalamic orexin (hypocretin) neurons express vesicular glutamate transporters VGLUT1 or VGLUT2. J Comp Neurol 2003; 465:593-603. [PMID: 12975818 DOI: 10.1002/cne.10860] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Initially recognized for their importance in control of appetite, orexins (also called hypocretins) are neuropeptides that are also involved in regulating sleep, arousal, and cardiovascular function. Loss of orexin appears to be the primary cause of narcolepsy. Cells expressing the orexins are restricted to a discrete region of the hypothalamus, but their terminal projections are widely distributed throughout the brain. With the diversity of function and broad distribution of orexin terminals, it is not known whether the orexin cells constitute a homogeneous population. Because orexins produce neuroexcitatory effects, we hypothesized that orexin-containing neurons are glutamatergic. In the present study we used digoxigenin-labeled cRNA probes for the vesicular glutamate transporters, VGLUT1 and VGLUT2, for in situ hybridization studies in combination with immunohistochemical detection of orexin cell bodies in the hypothalamus. In general, cells in the hypothalamus expressed low levels of the vesicular glutamate transporters relative to other areas of the forebrain, such as the cortex and thalamus. Light labeling for VGLUT2 mRNA was detected in about 50% of the orexin-immunoreactive neurons, and a much smaller percentage (approximately 13%) of orexin-immunoreactive cells was found to express VGLUT1. Despite the fact that intense labeling for GAD67 mRNA was found in a large number of cells throughout the hypothalamus, none of the orexin-immunoreactive cells was found to be GABAergic. These findings, showing that many of the orexin neurons are glutamatergic, are consistent with the neuroexcitatory effects of orexin but suggest that another neurochemical phenotype may define the remaining subset of orexin neurons.
Collapse
Affiliation(s)
- Diane L Rosin
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | |
Collapse
|
27
|
Li JL, Xiong KH, Dong YL, Fujiyama F, Kaneko T, Mizuno N. Vesicular glutamate transporters, VGluT1 and VGluT2, in the trigeminal ganglion neurons of the rat, with special reference to coexpression. J Comp Neurol 2003; 463:212-20. [PMID: 12815758 DOI: 10.1002/cne.10755] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vesicular glutamate transporters are responsible for glutamate transport into synaptic vesicles. In the present study, we examined immunohistochemically the expression of vesicular glutamate transporters, VGluT1 and VGluT2, in trigeminal ganglion neurons of the rat. Immunohistochemistry for VGluT1 and VGluT2 indicated that more than 80% of trigeminal ganglion neurons express VGluT1 and/or VGluT2 in their cell bodies. It also indicated that large and small trigeminal ganglion neurons express VGluT2 more frequently than VGluT1. Dual immunofluorescence histochemistry for VGluT1 and VGluT2 indicated that trigeminal ganglion neurons express VGluT2 more frequently than VGluT1 and that more than 80% of VGluT-expressing trigeminal ganglion neurons express VGluT1 and VGluT2. Many axon terminals in the superficial layers of the medullary dorsal horn also showed VGluT1 and VGluT2 immunoreactivities. Some of these axon terminals were confirmed to form the central core of the synaptic glomerulus. These results indicated that VGluT1 and VGluT2 are coexpressed in the cell bodies and axon terminals in most trigeminal ganglion neurons.
Collapse
Affiliation(s)
- Jin-Lian Li
- Department of Anatomy and K. K. Leung Brain Research Centre, The Fourth Military Medical University, Xi'an 710032, People's Republic of China
| | | | | | | | | | | |
Collapse
|
28
|
Ikemoto A, Bole DG, Ueda T. Glycolysis and glutamate accumulation into synaptic vesicles. Role of glyceraldehyde phosphate dehydrogenase and 3-phosphoglycerate kinase. J Biol Chem 2003; 278:5929-40. [PMID: 12488440 DOI: 10.1074/jbc.m211617200] [Citation(s) in RCA: 145] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Glucose is the major source of brain energy and is essential for maintaining normal brain and neuronal function. Hypoglycemia causes impaired synaptic transmission. This occurs even before significant reduction in global cellular ATP concentration, and relationships among glycolysis, ATP supply, and synaptic transmission are not well understood. We demonstrate that the glycolytic enzymes glyceraldehyde phosphate dehydrogenase (GAPDH) and 3-phosphoglycerate kinase (3-PGK) are enriched in synaptic vesicles, forming a functional complex, and that synaptic vesicles are capable of accumulating the excitatory neurotransmitter glutamate by harnessing ATP produced by vesicle-bound GAPDH/3-PGK at the expense of their substrates. The GAPDH inhibitor iodoacetate suppressed GAPDH/3-PGK-dependent, but not exogenous ATP-dependent, [(3)H]glutamate uptake into isolated synaptic vesicles. It also decreased vesicular [(3)H]glutamate content in the nerve ending preparation synaptosome; this decrease was reflected in reduction of depolarization-induced [(3)H]glutamate release. In contrast, oligomycin, a mitochondrial ATP synthase inhibitor, had minimal effect on any of these parameters. ADP at concentrations above 0.1 mm inhibited vesicular glutamate and dissipated membrane potential. This suggests that the coupled GAPDH/3-PGK system, which converts ADP to ATP, ensures maximal glutamate accumulation into presynaptic vesicles. Together, these observations provide insight into the essential nature of glycolysis in sustaining normal synaptic transmission.
Collapse
Affiliation(s)
- Atsushi Ikemoto
- Mental Health Research Institute, University of Michigan Medical School, Ann Arbor, Michigan 48109-0669, USA
| | | | | |
Collapse
|
29
|
Morimoto R, Hayashi M, Yatsushiro S, Otsuka M, Yamamoto A, Moriyama Y. Co-expression of vesicular glutamate transporters (VGLUT1 and VGLUT2) and their association with synaptic-like microvesicles in rat pinealocytes. J Neurochem 2003; 84:382-91. [PMID: 12559000 DOI: 10.1046/j.1471-4159.2003.01532.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A vesicular glutamate transporter (VGLUT) is responsible for the accumulation of l-glutamate in synaptic vesicles in glutamatergic neurons. Two isoforms, VGLUT1 and VGLUT2, have been identified, which are complementarily expressed in these neurons. Mammalian pinealocytes, endocrine cells for melatonin, are also glutamatergic in nature, accumulate l-glutamate in synaptic-like microvesicles (SLMVs), and secrete it through exocytosis. Although the storage of l-glutamate in SLMVs is mediated through a VGLUT, the molecular nature of the transporter is less understood. We recently observed that VGLUT2 is expressed in pinealocytes. In the present study, we show that pinealocytes also express VGLUT1. RT-PCR and northern blot analyses indicated expression of the VGLUT1 gene in pineal gland. Western blotting with specific antibodies against VGLUT1 indicated the presence of VGLUT1 in pineal gland. Indirect immunofluorescence microscopy with a section of pineal gland and cultured cells indicated that VGLUT1 and VGLUT2 are co-localized with process terminal regions of pinealocytes. Furthermore, immunoelectronmicroscopy as well as subcellular fractionation studies revealed that both VGLUT1 and VGLUT2 are specifically associated with SLMVs. These results indicate that both VGLUTs are responsible for storage of l-glutamate in SLMVs in pinealocytes. Pinealocytes are the first exception as to complementary expression of VGLUT1 and VGLUT2.
Collapse
Affiliation(s)
- Riyo Morimoto
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Okayama University, Japan.
| | | | | | | | | | | |
Collapse
|
30
|
Stornetta RL, Sevigny CP, Guyenet PG. Inspiratory augmenting bulbospinal neurons express both glutamatergic and enkephalinergic phenotypes. J Comp Neurol 2003; 455:113-24. [PMID: 12455000 DOI: 10.1002/cne.10486] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many of the inspiratory augmenting (I-AUG) neurons of the rostral ventral respiratory group (rVRG) are premotor neurons that excite phrenic motor neurons during inspiration, probably by releasing glutamate. In the present study, we demonstrate that these neurons are indeed glutamatergic, in that their cell bodies contain vesicular glutamate transporter-2 (VGLUT2) mRNA and spinal terminals from neurons in the region of the rVRG contain VGLUT2 protein. We also demonstrate by using parallel in situ hybridization and immunocytochemical evidence that most rVRG inspiratory premotor neurons are enkephalinergic. After iontophoretic deposits of biotinylated dextran amine (BDA) in the area of the rVRG, many BDA-labeled terminals in the ventral horn of cervical spinal cord (C4-C5) were immunoreactive for enkephalin and VGLUT2. Injections of Fluoro-Gold amidst phrenic motor neurons in C4-C5 labeled neurons in the area of the rVRG that contained both VGLUT2 mRNA and preproenkephalin (PPE) mRNA as revealed by double in situ hybridization. Thirty-eight bulbospinal I-AUG neurons were recorded in the rVRG and filled with biotinamide by using the juxtacellular labeling technique. Every biotinamide-filled cell tested was positively labeled for VGLUT2 mRNA (n = 14), and most of the cells tested in a separate population exhibited PPE mRNA (16/18). We conclude that most of the phrenic inspiratory premotor neurons of the rVRG are glutamatergic neurons that may also release enkephalins.
Collapse
Affiliation(s)
- Ruth L Stornetta
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia 22908-0735, USA.
| | | | | |
Collapse
|
31
|
Bole DG, Hirata K, Ueda T. Prolonged depolarization of rat cerebral synaptosomes leads to an increase in vesicular glutamate content. Neurosci Lett 2002; 322:17-20. [PMID: 11958833 DOI: 10.1016/s0304-3940(02)00105-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Glutamate accumulation into synaptic vesicles is a vital step in glutamate synaptic transmission. In this study, we have explored the possibility that vesicular glutamate storage may be subject to some regulation. Synaptosomes were depolarized and subjected to [3H] glutamate under non-depolarizing conditions, and vesicular [3H] glutamate content was determined by a filter-based assay. We present evidence here that prolonged depolarization of synaptosomes leads to an increase in vesicular glutamate content. Induction of this enhanced state is time- and temperature-dependent. The enhanced state has two components, one readily reversible and the other long-lasting. The up-regulation of glutamate storage capacity could lead to an increase in quantal size and play a role in modulation of glutamate transmission efficiency.
Collapse
Affiliation(s)
- David G Bole
- Mental Health Research Institute, University of Michigan Medical School, 205 Zina Pitcher Place, Ann Arbor, MI 48109-0720, USA
| | | | | |
Collapse
|
32
|
Stornetta RL, Sevigny CP, Schreihofer AM, Rosin DL, Guyenet PG. Vesicular glutamate transporter DNPI/VGLUT2 is expressed by both C1 adrenergic and nonaminergic presympathetic vasomotor neurons of the rat medulla. J Comp Neurol 2002; 444:207-20. [PMID: 11840475 DOI: 10.1002/cne.10142] [Citation(s) in RCA: 154] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The main source of excitatory drive to the sympathetic preganglionic neurons that control blood pressure is from neurons located in the rostral ventrolateral medulla (RVLM). This monosynaptic input includes adrenergic (C1), peptidergic, and noncatecholaminergic neurons. Some of the cells in this pathway are suspected to be glutamatergic, but conclusive evidence is lacking. In the present study we sought to determine whether these presympathetic neurons express the vesicular glutamate transporter BNPI/VGLUT1 or the closely related gene DNPI, the rat homolog of the mouse vesicular glutamate transporter VGLUT2. Both BNPI/VGLUT1 and DNPI/VGLUT2 mRNAs were detected in the medulla oblongata by in situ hybridization, but only DNPI/VGLUT2 mRNA was present in the RVLM. Moreover, BNPI immunoreactivity was absent from the thoracic spinal cord lateral horn. DNPI/VGLUT2 mRNA was present in many medullary cells retrogradely labeled with Fluoro-Gold from the spinal cord (T2; four rats). Within the RVLM, 79% of the bulbospinal C1 cells contained DNPI/VGLUT2 mRNA. Bulbospinal noradrenergic A5 neurons did not contain DNPI/VGLUT2 mRNA. The RVLM of six unanesthetized rats subjected to 2 hours of hydralazine-induced hypotension contained tenfold more c-Fos-ir DNPI/VGLUT2 neurons than that of six saline-treated controls. c-Fos-ir DNPI/VGLUT2 neurons included C1 and non-C1 neurons (3:2 ratio). In seven barbiturate-anesthetized rats, 16 vasomotor presympathetic neurons were filled with biotinamide and analyzed for the presence of tyrosine hydroxylase immunoreactivity and/or DNPI/VGLUT2 mRNA. Biotinamide-labeled neurons included C1 and non-C1 cells. Most non-C1 (9/10) and C1 presympathetic cells (5/6) contained DNPI/VGLUT2 mRNA. In conclusion, DNPI/VGLUT2 is expressed by most blood pressure-regulating presympathetic cells of the RVLM. The data suggest that these neurons may be glutamatergic and that the C1 adrenergic phenotype is one of several secondary phenotypes that are differentially expressed by subgroups of these cells.
Collapse
Affiliation(s)
- Ruth L Stornetta
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia 22908-0735, USA.
| | | | | | | | | |
Collapse
|
33
|
Stornetta RL, Sevigny CP, Guyenet PG. Vesicular glutamate transporter DNPI/VGLUT2 mRNA is present in C1 and several other groups of brainstem catecholaminergic neurons. J Comp Neurol 2002; 444:191-206. [PMID: 11840474 DOI: 10.1002/cne.10141] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mouse glutamate vesicular transporter VGLUT2 has recently been characterized. The rat homolog of VGLUT2, differentiation-associated Na(+)/P(i) cotransporter (DNPI), was examined using a digoxigenin-labeled DNPI/VGLUT2 cRNA probe in the present study to determine which, if any, of the various groups of pontine or medullary monoaminergic neurons express DNPI/VGLUT2 mRNA and, thus, are potentially glutamatergic. DNPI/VGLUT2 mRNA was widely distributed within the brainstem and seemed exclusively neuronal. By using a double in situ hybridization method, the presence of the mRNA for DNPI/VGLUT2 and glutamic acid decarboxylase (GAD)-67 was mutually exclusive. By combining DNPI/VGLUT2 mRNA detection and conventional immunohistochemistry, DNPI/VGLUT2 mRNA was undetectable in lower brainstem cholinergic and serotonergic cells, but it was present in several tyrosine hydroxylase-immunoreactive (TH-ir) cell groups. DNPI/VGLUT2 mRNA was detected in most of the adrenergic neurons of the C1, C2, and C3 groups (75-80% of TH-ir neurons), in the A2 noradrenergic group (80%), and in vast numbers of area postrema cells. Within the A1 region, many fewer TH-ir cells contained DNPI/VGLUT2 (16%). Finally, DNPI/VGLUT2 mRNA was undetectable in the pontine noradrenergic cell groups (A5 and A6/locus coeruleus). In conclusion, the general pattern of DNPI/VGLUT2 expression and its exclusion from GABAergic, cholinergic, and serotonergic neurons supports the notion that DNPI/VGLUT2 mRNA identifies a subset of glutamatergic neurons in the lower brainstem. Within this region several catecholaminergic cell groups appear to be glutamatergic, including but not limited to the adrenergic cell groups C1-C3. Based on the present evidence, the noradrenergic cell groups of the pons (A5 and A6) do not contain either known vesicular glutamate transporter and are most likely not glutamatergic.
Collapse
Affiliation(s)
- Ruth L Stornetta
- Department of Pharmacology, University of Virginia Health System, Charlottesville, Virginia 22908-0735, USA.
| | | | | |
Collapse
|
34
|
Staley KJ. Diuretics as Antiepileptic Drugs: Should We Go with the Flow? Epilepsy Curr 2002; 2:35-38. [PMID: 15309160 PMCID: PMC320967 DOI: 10.1111/j.1535-7597.2002.00018.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Recent epidemiological and experimental studies have suggested that certain diuretics may have significant anticonvulsant actions. Potential anticonvulsant mechanisms are discussed in light of the effects of these diuretics on electrolyte balance and synaptic signaling.
Collapse
Affiliation(s)
- Kevin J. Staley
- />Department of Neurology, University of Colorado, Denver, Colorado
| |
Collapse
|
35
|
Abstract
Recent epidemiological and experimental studies have suggested that certain diuretics may have significant anticonvulsant actions. Potential anticonvulsant mechanisms are discussed in light of the effects of these diuretics on electrolyte balance and synaptic signaling.
Collapse
Affiliation(s)
- Kevin J. Staley
- Department of Neurology, University of Colorado, Denver, Colorado
| |
Collapse
|
36
|
Identification of the differentiation-associated Na+/PI transporter as a novel vesicular glutamate transporter expressed in a distinct set of glutamatergic synapses. J Neurosci 2002. [PMID: 11756497 DOI: 10.1523/jneurosci.22-01-00142.2002] [Citation(s) in RCA: 329] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Glutamate transport into synaptic vesicles is a prerequisite for its regulated neurosecretion. Here we functionally identify a second isoform of the vesicular glutamate transporter (VGLUT2) that was previously identified as a plasma membrane Na+-dependent inorganic phosphate transporter (differentiation-associated Na+/P(I) transporter). Studies using intracellular vesicles from transiently transfected PC12 cells indicate that uptake by VGLUT2 is highly selective for glutamate, is H+ dependent, and requires Cl- ion. Both the vesicular membrane potential (Deltapsi) and the proton gradient (DeltapH) are important driving forces for vesicular glutamate accumulation under physiological Cl- concentrations. Using an antibody specific for VGLUT2, we also find that this protein is enriched on synaptic vesicles and selective for a distinct class of glutamatergic nerve terminals. The pathway-specific, complementary expression of two different vesicular glutamate transporters suggests functional diversity in the regulation of vesicular release at excitatory synapses. Together, the two isoforms may account for the uptake of glutamate by synaptic vesicles from all central glutamatergic neurons.
Collapse
|
37
|
Hayashi M, Otsuka M, Morimoto R, Hirota S, Yatsushiro S, Takeda J, Yamamoto A, Moriyama Y. Differentiation-associated Na+-dependent inorganic phosphate cotransporter (DNPI) is a vesicular glutamate transporter in endocrine glutamatergic systems. J Biol Chem 2001; 276:43400-6. [PMID: 11551935 DOI: 10.1074/jbc.m106244200] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Vesicular glutamate transporter is present in neuronal synaptic vesicles and endocrine synaptic-like microvesicles and is responsible for vesicular storage of L-glutamate. A brain-specific Na(+)-dependent inorganic phosphate transporter (BNPI) functions as a vesicular glutamate transporter in synaptic vesicles, and the expression of this BNPI defines the glutamatergic phenotype in the central nervous system (Bellocchio, E. E., Reimer, R. J., Fremeau, R. T., Jr., and Edwards, R. H. (2000) Science 289, 957-960; Takamori, S., Rhee, J. S., Rosenmund, C., and Jahn, R. (2000) Nature 407, 189-194). However, since not all glutamatergic neurons contain BNPI, an additional transporter(s) responsible for vesicular glutamate uptake has been postulated. Here we report that differentiation-associated Na(+)-dependent inorganic phosphate cotransporter (DNPI), an isoform of BNPI (Aihara, Y., Mashima, H., Onda, H., Hisano, S., Kasuya, H., Hori, T., Yamada, S., Tomura, H., Yamada, Y., Inoue, I., Kojima, I., and Takeda, J. (2000) J. Neurochem. 74, 2622-2625), also transports L-glutamate at the expense of an electrochemical gradient of protons established by the vacuolar proton pump when expressed in COS7 cells. Molecular, biological, and immunohistochemical studies have indicated that besides its presence in neuronal cells DNPI is preferentially expressed in mammalian pinealocytes, alphaTC6 cells, clonal pancreatic alpha cells, and alpha cells of Langerhans islets, these cells being proven to secrete L-glutamate through Ca(2+)-dependent regulated exocytosis followed by its vesicular storage. Pancreatic polypeptide-secreting F cells of Langerhans islets also expressed DNPI. These results constitute evidence that DNPI functions as another vesicular transporter in glutamatergic endocrine cells as well as in neurons.
Collapse
Affiliation(s)
- M Hayashi
- Department of Biochemistry, Faculty of Pharmaceutical Sciences, Okayama University, Okayama 700-8530, Japan
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Fremeau RT, Troyer MD, Pahner I, Nygaard GO, Tran CH, Reimer RJ, Bellocchio EE, Fortin D, Storm-Mathisen J, Edwards RH. The expression of vesicular glutamate transporters defines two classes of excitatory synapse. Neuron 2001; 31:247-60. [PMID: 11502256 DOI: 10.1016/s0896-6273(01)00344-0] [Citation(s) in RCA: 984] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The quantal release of glutamate depends on its transport into synaptic vesicles. Recent work has shown that a protein previously implicated in the uptake of inorganic phosphate across the plasma membrane catalyzes glutamate uptake by synaptic vesicles. However, only a subset of glutamate neurons expresses this vesicular glutamate transporter (VGLUT1). We now report that excitatory neurons lacking VGLUT1 express a closely related protein that has also been implicated in phosphate transport. Like VGLUT1, this protein localizes to synaptic vesicles and functions as a vesicular glutamate transporter (VGLUT2). The complementary expression of VGLUT1 and 2 defines two distinct classes of excitatory synapse.
Collapse
Affiliation(s)
- R T Fremeau
- Department of Neurology, Graduate Program in Neuroscience, UCSF School of Medicine, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Somogyi J, Llewellyn-Smith IJ. Patterns of colocalization of GABA, glutamate and glycine immunoreactivities in terminals that synapse on dendrites of noradrenergic neurons in rat locus coeruleus. Eur J Neurosci 2001; 14:219-28. [PMID: 11553275 DOI: 10.1046/j.0953-816x.2001.01638.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Amino acid transmitters play a key role in regulating the activity of noradrenergic neurons in the locus coeruleus. We investigated the anatomical substrate for this regulation by quantifying immunoreactivity for GABA, glutamate and glycine in terminals that contacted the dendrites of tyrosine hydroxylase-immunoreactive principal neurons in rat locus coeruleus. Pre-embedding peroxidase immunocytochemistry was used to detect tyrosine hydroxylase-immunoreactivity in Vibratome sections of tissue perfused with 2.5% glutaraldehyde. GABA, glutamate and glycine were localized with postembedding immunogold labelling. Gold particle densities over terminals were measured in three semiserial ultrathin sections, each reacted for a different amino acid. More than 90% (range among rats, 89%-95%) of the terminals analyzed (n = 288) were immunoreactive for at least one amino acid. A high proportion (39%-49%) were positive for two or three amino acids. About two-thirds (60%-69%) of the boutons contained GABA, of which more than half (51%-55%) also contained glycine. More than one-third (36%-38%) of the terminals were positive for glycine. Terminals immunoreactive for glycine alone were rare (0%-2%). About one-third of the terminals showed glutamate-immunoreactivity (32%-37%). GABA and/or glycine occurred in one-fifth to one-third of these. These results show that amino acid-immunoreactivity is present in almost all of the terminals that synapse on tyrosine hydroxylase-positive dendrites in locus coeruleus. Glutamate provides a major excitatory input. The almost complete colocalization of glycine with GABA suggests that the inhibitory input to locus coeruleus is predominantly GABAergic with a contribution from glycine in about half of the GABAergic boutons.
Collapse
Affiliation(s)
- J Somogyi
- Cardiovascular Neuroscience Group, Cardiovascular Medicine and Centre for Neuroscience, Flinders University, Bedford Park, SA 5042, Australia.
| | | |
Collapse
|
40
|
Balcar VJ, Takamoto A, Yoneda Y. Neurochemistry of L-Glutamate Transport in the CNS: A Review of Thirty Years of Progress. ACTA ACUST UNITED AC 2001. [DOI: 10.1135/cccc20011315] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The review highlights the landmark studies leading from the discovery and initial characterization of the Na+-dependent "high affinity" uptake in the mammalian brain to the cloning of individual transporters and the subsequent expansion of the field into the realm of molecular biology. When the data and hypotheses from 1970's are confronted with the recent developments in the field, we can conclude that the suggestions made nearly thirty years ago were essentially correct: the uptake, mediated by an active transport into neurons and glial cells, serves to control the extracellular concentrations of L-glutamate and prevents the neurotoxicity. The modern techniques of molecular biology may have provided additional data on the nature and location of the transporters but the classical neurochemical approach, using structural analogues of glutamate designed as specific inhibitors or substrates for glutamate transport, has been crucial for the investigations of particular roles that glutamate transport might play in health and disease. Analysis of recent structure/activity data presented in this review has yielded a novel insight into the pharmacological characteristics of L-glutamate transport, suggesting existence of additional heterogeneity in the system, beyond that so far discovered by molecular genetics. More compounds that specifically interact with individual glutamate transporters are urgently needed for more detailed investigations of neurochemical characteristics of glutamatergic transport and its integration into the glutamatergic synapses in the central nervous system. A review with 162 references.
Collapse
|