1
|
Corty M, Arlt FA, Borowski K, Wernick S, Scharf M, Piecha FA, Steen C, Oehler P, Buthut M, Krill M, Zöllner JP, Krafft T, Markewitz R, Radzimski C, Denno Y, Probst C, Teegen B, Grefkes C, Knake S, Witt K, Finke C, Hagemann G, Becker J, Kraemer M, Witte T, Komorowski L, Wandinger KP, Prüss H, Melzer N, Miske R. Anti-VGLUT2 autoantibodies in neurological diseases. Brain Behav Immun 2025; 129:470-484. [PMID: 40494504 DOI: 10.1016/j.bbi.2025.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 06/04/2025] [Accepted: 06/07/2025] [Indexed: 06/29/2025] Open
Abstract
Autoantibodies are important biomarkers for the diagnosis of autoimmune diseases that help to determine treatment strategies and to understand disease pathology. Despite the increasing numbers of neuronal autoantibody discoveries, there are still patients presenting with neurological autoimmune diseases and so far uncharacterized autoantibodies. Between 12/2016 and 06/2024, we collected sera of 314 patients with a distinct uncharacterized IgG pattern in neuronal tissue indirect immunofluorescence assay (IIFA). By immunoprecipitation and mass spectrometry we identified vesicular glutamate transporter 2 (VGLUT2) as the autoantibody target and confirmed it in sera of 285/314 patients by recombinant IIFA with VGLUT2-expressing HEK293 cells, competitive inhibition assays and colocalization studies with a commercial antibody. The main diagnoses available of 87/285 patients (mean age 58, range 1-92) were encephalitis 25/87, dementia/cognitive impairment 17/87 and polyneuropathy 16/87. Detailed clinical data of 18 patients were collected retrospectively. The major symptoms of those index patients (mean age 56, range 2-78) involved cognitive changes (10/18) including memory impairment, aphasia and disorientation as well as sensorimotor disturbances (9/18) and gait abnormalities (9/18), accompanied by visual impairments (8/18). (Poly)neuropathy was observed in 10/18 cases. The majority of the anti-VGLUT2 index patients had coexisting diabetes mellitus type 2 or other chronic multisystem disorders. In 8/12 index patients, immunotherapy had beneficial effects, although only slight improvements could be obtained in most of the cases. All 17 analyzed index patient sera recognized a cytoplasmic epitope between amino acid 520-564 of VGLUT2, determined by immunoblot with recombinant antigen fragments. Anti-VGLUT2 autoantibody-associated neurological diseases may represent a new type of autoimmune disorders that might benefit from immunomodulatory treatment and predominantly manifests with encephalitis, cognitive deficits and neuropathy.
Collapse
Affiliation(s)
- Merle Corty
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Groß Grönau, Germany
| | - Friederike A Arlt
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Kathrin Borowski
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Groß Grönau, Germany
| | - Stephanie Wernick
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Madeleine Scharf
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | | | - Claudia Steen
- St. Joseph Hospital Berlin, Department for sick children, Berlin, Germany
| | - Philipp Oehler
- St. Joseph Hospital Berlin, Department for sick children, Berlin, Germany
| | - Maria Buthut
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany; Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Moritz Krill
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Johann-Philipp Zöllner
- Goethe University Frankfurt, University Hospital, Department of Neurology, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Tammo Krafft
- Evangelic Hospital Oldenburg, Steinweg 13-17, 25122 Oldenburg, Germany
| | - Robert Markewitz
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Christiane Radzimski
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Yvonne Denno
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Christian Probst
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. h.c. (RCH) Dr. med. Winfried Stöcker, Groß Grönau, Germany
| | - Christian Grefkes
- Goethe University Frankfurt, University Hospital, Department of Neurology, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Susanne Knake
- Philipps-Universität Marburg, Fachbereich Medizin, Klinik für Neurologie, Epilepsiezentrum Hessen, Marburg, Germany
| | - Karsten Witt
- Evangelic Hospital Oldenburg, Steinweg 13-17, 25122 Oldenburg, Germany; Department of Neurology, University Oldenburg, Steinweg 13-17, 26122 Oldenburg, Germany; Research Center of Neurosensory Science, Carl Von Ossietzky University of Oldenburg, Oldenburg, Germany
| | - Carsten Finke
- Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Georg Hagemann
- Department of Neurology, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Jana Becker
- Department of Neurology, Alfried Krupp Hospital, Alfried-Krupp-Strasse 21, 45130 Essen, Germany
| | - Markus Kraemer
- Department of Neurology, Alfried Krupp Hospital, Alfried-Krupp-Strasse 21, 45130 Essen, Germany; Department of Neurology, Medical Faculty and University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Torsten Witte
- Department of Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Lars Komorowski
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany
| | - Klaus-Peter Wandinger
- Institute of Clinical Chemistry, University Hospital Schleswig-Holstein, Kiel/Lübeck, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Berlin, Berlin, Germany
| | - Nico Melzer
- Department of Neurology, Medical Faculty and University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Ramona Miske
- Institute for Experimental Immunology, affiliated with EUROIMMUN Medizinische Labordiagnostika AG, Seekamp 31, 23560 Luebeck, Germany.
| |
Collapse
|
2
|
Li Y, Gong L, Wu J, Hung W, Zhen M, Gao S. UBR-1 deficiency leads to ivermectin resistance in Caenorhabditis elegans. eLife 2025; 13:RP103718. [PMID: 40167441 PMCID: PMC11961118 DOI: 10.7554/elife.103718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Resistance to anthelmintics, particularly the macrocyclic lactone ivermectin (IVM), presents a substantial global challenge for parasite control. We found that the functional loss of an evolutionarily conserved E3 ubiquitin ligase, UBR-1, leads to IVM resistance in Caenorhabditis elegans. Multiple IVM-inhibiting activities, including viability, body size, pharyngeal pumping, and locomotion, were significantly ameliorated in various ubr-1 mutants. Interestingly, exogenous application of glutamate induces IVM resistance in wild-type animals. The sensitivity of all IVM-affected phenotypes of ubr-1 is restored by eliminating proteins associated with glutamate metabolism or signaling: GOT-1, a transaminase that converts aspartate to glutamate, and EAT-4, a vesicular glutamate transporter. We demonstrated that IVM-targeted GluCls (glutamate-gated chloride channels) are downregulated and that the IVM-mediated inhibition of serotonin-activated pharynx Ca2+ activity is diminished in ubr-1. Additionally, enhancing glutamate uptake in ubr-1 mutants through ceftriaxone completely restored their IVM sensitivity. Therefore, UBR-1 deficiency-mediated aberrant glutamate signaling leads to ivermectin resistance in C. elegans.
Collapse
Affiliation(s)
- Yi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Long Gong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Jing Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| | - Wesley Hung
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of TorontoTorontoCanada
| | - Mei Zhen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of TorontoTorontoCanada
| | - Shangbang Gao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
3
|
Zhao Y, Zhang Y, Feng J, He Z, Li T. Codon Usage Bias: A Potential Factor Affecting VGLUT Developmental Expression and Protein Evolution. Mol Neurobiol 2025; 62:3508-3522. [PMID: 39305444 DOI: 10.1007/s12035-024-04426-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 08/05/2024] [Indexed: 02/04/2025]
Abstract
More and more attention has been paid to the role of synonymous substitution in evolution, in which codon usage preference can affect gene expression distribution and protein structure and function. Vesicular glutamate transporter (VGLUT) consists of three isoforms, among which VGLUT3 is significantly different from other VGLUTs in functional importance, expression level, and distribution range, whose reason is still unclear. This study sought to analyze the role of codon preference in VGLUT differentiation. To conduct an evolutionary analysis of the three VGLUTs, this paper uses bioinformatics research methods to analyze the coding sequences of the three VGLUTs in different species and compare the codon usage patterns. Furthermore, the differences among the three VGLUTs were analyzed by combining functional importance, expression level, distribution range, gene structure, protein relationship network, expression at specific developmental stages, and phylogenetic tree, and the influence of codon usage pattern was explored. The results showed that the VGLUT with greater codon preference had less functional importance, lower expression levels, more peripheral distribution away from the CNS, smaller exon density of gene, less conserved and farther away from the CDS region miRNA regulatory sites, simpler and less tight protein interaction networks, delayed developmental expression, and more distant evolutionary relationships. Codon usage preference is a potential factor affecting VGLUT developmental expression and protein evolution.
Collapse
Affiliation(s)
- Yiran Zhao
- College of Life Sciences, Yunlong District, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou City, Jiangsu, 221000, China
| | - Yu Zhang
- College of Life Sciences, Yunlong District, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou City, Jiangsu, 221000, China
| | - Jiaxing Feng
- College of Life Sciences, Yunlong District, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou City, Jiangsu, 221000, China
| | - Zixian He
- College of Life Sciences, Yunlong District, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou City, Jiangsu, 221000, China
| | - Ting Li
- College of Life Sciences, Yunlong District, Xuzhou Medical University, No. 209, Tongshan Road, Xuzhou City, Jiangsu, 221000, China.
| |
Collapse
|
4
|
Galli A, Moretti S, Dule N, Di Cairano ES, Castagna M, Marciani P, Battaglia C, Bertuzzi F, Fiorina P, Pastore I, La Rosa S, Davalli A, Folli F, Perego C. Hyperglycemia impairs EAAT2 glutamate transporter trafficking and glutamate clearance in islets of Langerhans: implications for type 2 diabetes pathogenesis and treatment. Am J Physiol Endocrinol Metab 2024; 327:E27-E41. [PMID: 38690938 PMCID: PMC11390119 DOI: 10.1152/ajpendo.00069.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/18/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Abstract
Pancreatic endocrine cells employ a sophisticated system of paracrine and autocrine signals to synchronize their activities, including glutamate, which controls hormone release and β-cell viability by acting on glutamate receptors expressed by endocrine cells. We here investigate whether alteration of the excitatory amino acid transporter 2 (EAAT2), the major glutamate clearance system in the islet, may occur in type 2 diabetes mellitus and contribute to β-cell dysfunction. Increased EAAT2 intracellular localization was evident in islets of Langerhans from T2DM subjects as compared with healthy control subjects, despite similar expression levels. Chronic treatment of islets from healthy donors with high-glucose concentrations led to the transporter internalization in vesicular compartments and reduced [H3]-d-glutamate uptake (65 ± 5% inhibition), phenocopying the findings in T2DM pancreatic sections. The transporter relocalization was associated with decreased Akt phosphorylation protein levels, suggesting an involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the process. In line with this, PI3K inhibition by a 100-µM LY294002 treatment in human and clonal β-cells caused the transporter relocalization in intracellular compartments and significantly reduced the glutamate uptake compared to control conditions, suggesting that hyperglycemia changes the trafficking of the transporter to the plasma membrane. Upregulation of the glutamate transporter upon treatment with the antibiotic ceftriaxone rescued hyperglycemia-induced β-cells dysfunction and death. Our data underscore the significance of EAAT2 in regulating islet physiology and provide a rationale for potential therapeutic targeting of this transporter to preserve β-cell survival and function in diabetes.NEW & NOTEWORTHY The glutamate transporter SLC1A2/excitatory amino acid transporter 2 (EAAT2) is expressed on the plasma membrane of pancreatic β-cells and controls islet glutamate clearance and β-cells survival. We found that the EAAT2 membrane expression is lost in the islets of Langerhans from type 2 diabetes mellitus (T2DM) patients due to hyperglycemia-induced downregulation of the phosphoinositide 3-kinase/Akt pathway and modification of its intracellular trafficking. Pharmacological rescue of EAAT2 expression prevents β-cell dysfunction and death, suggesting EAAT2 as a new potential target of intervention in T2DM.
Collapse
Affiliation(s)
- Alessandra Galli
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Nevia Dule
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Eliana Sara Di Cairano
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Cristina Battaglia
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | | | - Paolo Fiorina
- Department of Biomedical and Clinical Sciences "L. Sacco,"Università degli Studi di Milano, Milan, Italy
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Unit, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Stefano La Rosa
- Unit of Pathology, Department of Oncology, ASST Sette Laghi, Varese, Italy
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
| | - Alberto Davalli
- Diabetes and Endocrinology Unit, Department of Internal Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Carla Perego
- Laboratory of Molecular and Cellular Physiology, Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Teng L, Qin Q, Zhou Z, Zhou F, Cao C, Yang J, Ding J. Glutamate secretion by embryonic stem cells as an autocrine signal to promote proliferation. Sci Rep 2023; 13:19069. [PMID: 37925518 PMCID: PMC10625544 DOI: 10.1038/s41598-023-46477-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/01/2023] [Indexed: 11/06/2023] Open
Abstract
Glutamate, the major excitatory neurotransmitter in the central nervous system, has also been found to play a role in embryonic stem (ES) cells. However, the exact mechanism and function of glutamatergic signaling in ES cells remain poorly understood. In this study, we identified a glutamatergic transmission circuit in ES cells that operates through an autocrine mechanism and regulates cell proliferation. We performed biological analyses to identify the key components involved in glutamate biosynthesis, packaging for secretion, reaction, and reuptake in ES cells, including glutaminase, vesicular glutamate transporter, glutamate N-methyl-D-aspartate (NMDA) receptor, and cell membrane excitatory amino-acid transporter (EAAT). We directly quantified the released glutamate signal using microdialysis-high performance liquid chromatography-tandem mass spectrometry (MD-HPLC-MS-MS). Pharmacological inhibition of endogenous glutamate release and the resulting tonic activation of NMDA receptors significantly affected ES cell proliferation, suggesting that ES cells establish a glutamatergic autocrine niche via releasing and responding to the transmitter for their own regulation.
Collapse
Affiliation(s)
- Lin Teng
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Qin Qin
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Ziyi Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Fei Zhou
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Chunyu Cao
- College of Basic Medical Sciences, Hubei Key Laboratory of Tumor Microencironment and Immunotherapy, China Three Gorges University, Yichang, 443000, Hubei, China
| | - Jian Yang
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China
| | - Jiawang Ding
- Department of Cardiology, Yichang Central People's Hospital/The First College of Clinical Medical Sciences, China Three Gorges University, No. 183 Yiling Road, Yichang, 443003, Hubei, China.
- Institute of Cardiovascular Disease, China Three Gorges University, Yichang, 443003, Hubei, China.
| |
Collapse
|
6
|
Lenhart A, Kennedy RT. Evaluation of Surface Treatments of PDMS Microfluidic Devices for Improving Small-Molecule Recovery with Application to Monitoring Metabolites Secreted from Islets of Langerhans. ACS MEASUREMENT SCIENCE AU 2023; 3:380-389. [PMID: 37868359 PMCID: PMC10588933 DOI: 10.1021/acsmeasuresciau.3c00025] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 10/24/2023]
Abstract
Microfluidic devices are becoming an important tool for bioanalysis with applications including studying cell secretion, cell growth, and drug delivery. Small molecules such as drugs, cell products, or nutrients may partition into polydimethylsiloxane (PDMS), a commonly used material for microfluidic devices, potentially leading to poor recovery or inaccurate delivery of such chemicals. To decrease small-molecule partitioning, surface and bulk PDMS treatments have been developed; however, these have been tested on few analytes, or their biocompatibility are unknown. Studies often focus on one analyte, whereas a diversity of chemicals are of interest and possibly affected. In this study, 11 device treatments are tested and applied to 21 biologically relevant small molecules with a variety of chemical structures. Device treatments are characterized using water contact angle measurements and evaluated by measuring recovery of the 21 target analytes using liquid chromatography-mass spectrometry. 1,5-Dimethyl-1,5-diazaundecamethylene polymethobromide (polybrene), a positively charged polymer, produced the least hydrophilic surface and was found to provide the best recovery with most of the analytes having >50% recovery and up to 92% recovery; however, recovery varied by analyte highlighting the importance of analyte diversity rather than targeting a single analyte in evaluating treatments. A polybrene-treated device was applied to investigate secretion from pancreatic islets, which are micro-organs involved in glucose homeostasis and diabetes. Islets secrete small molecules that have been shown to modulate the secretion of islets' main functional products, glucose-regulating hormones. The polybrene treatment enabled the detection of 20 target analytes from islets-on-chip during isosmotic and hypo-osmotic glucose perfusions and resulted in detection of more significant secretion changes compared to untreated PDMS.
Collapse
Affiliation(s)
- Ashley
E. Lenhart
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Robert T. Kennedy
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
7
|
Monitoring hormone and small molecule secretion dynamics from islets-on-chip. Anal Bioanal Chem 2023; 415:533-544. [PMID: 36459167 DOI: 10.1007/s00216-022-04460-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022]
Abstract
Tissue functions such as hormone secretion involve the interplay of multiple chemical signals and metabolic processes over time. Measuring the different components involved is useful in unraveling the interactions, but often requires use of multiple analytical techniques. The challenge of measuring the necessary components with temporal resolution is greater when tissue samples are limited. Here, an accessible microfluidic platform compatible with multiple measurement techniques to monitor cell secretions has been developed. The platform is applied to islets of Langerhans, micro-organs involved in glucose homeostasis and diabetes. The device houses 1 to 8 islets and the perfusion fluid can be controlled to change conditions, e.g., glucose concentration, in seconds. Samples are collected in fractions and split for offline analysis. The device is paired with a scaled-down immunoassay, AlphaLISA, for hormone quantification and liquid chromatography-mass spectrometry for small molecule quantification to study secretion dynamics. The combined system allows the first simultaneous measurement of insulin, glucagon, biogenic amines, and amino acids from islet secretions. The combined measurements revealed correlation in secretion events and differences in timing of release between hormones and biogenic amines and amino acids. These efforts decreased the number of islets required compared to standard approaches, thus decreasing necessary animal use, reagent use, and cost, while increasing information content achievable from one sample. The microfluidic device is a suitable platform for in-depth characterization of secretion from small tissue samples.
Collapse
|
8
|
Panzer JK, Tamayo A, Caicedo A. Restoring glutamate receptor signaling in pancreatic alpha cells rescues glucagon responses in type 1 diabetes. Cell Rep 2022; 41:111792. [PMID: 36516761 DOI: 10.1016/j.celrep.2022.111792] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 05/19/2022] [Accepted: 11/16/2022] [Indexed: 12/15/2022] Open
Abstract
Glucagon secretion from pancreatic alpha cells is crucial to prevent hypoglycemia. People with type 1 diabetes lose this glucoregulatory mechanism and are susceptible to dangerous hypoglycemia for reasons still unclear. Here we determine that alpha cells in living pancreas slices from donors with type 1 diabetes do not mount an adequate glucagon response and cannot activate the positive autocrine feedback mediated by AMPA/kainate glutamate receptors. This feedback is required to elicit full glucagon responses in the healthy state. Reactivating residual AMPA/kainate receptor function with positive allosteric modulators restores glucagon secretion in human slices from donors with type 1 diabetes as well as glucose counterregulation in non-obese diabetic mice. Our study thus identifies a defect in autocrine signaling that contributes to alpha cell failure. The use of positive allosteric modulators of AMPA/kainate receptors overcomes this deficiency and prevents hypoglycemia, an effect that could be used to improve the management of diabetes.
Collapse
Affiliation(s)
- Julia K Panzer
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Alejandro Tamayo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alejandro Caicedo
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
9
|
Zmazek J, Grubelnik V, Markovič R, Marhl M. Modeling the Amino Acid Effect on Glucagon Secretion from Pancreatic Alpha Cells. Metabolites 2022; 12:metabo12040348. [PMID: 35448534 PMCID: PMC9028923 DOI: 10.3390/metabo12040348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 Diabetes Mellitus (T2DM) is a burdensome problem in modern society, and intensive research is focused on better understanding the underlying cellular mechanisms of hormone secretion for blood glucose regulation. T2DM is a bi-hormonal disease, and in addition to 100 years of increasing knowledge about the importance of insulin, the second hormone glucagon, secreted by pancreatic alpha cells, is becoming increasingly important. We have developed a mathematical model for glucagon secretion that incorporates all major metabolic processes of glucose, fatty acids, and glutamine as the most abundant postprandial amino acid in blood. In addition, we consider cAMP signaling in alpha cells. The model predictions quantitatively estimate the relative importance of specific metabolic and signaling pathways and particularly emphasize the important role of glutamine in promoting glucagon secretion, which is in good agreement with known experimental data.
Collapse
Affiliation(s)
- Jan Zmazek
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
| | - Vladimir Grubelnik
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Rene Markovič
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Electrical Engineering and Computer Science, University of Maribor, 2000 Maribor, Slovenia;
| | - Marko Marhl
- Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia; (J.Z.); (R.M.)
- Faculty of Education, University of Maribor, 2000 Maribor, Slovenia
- Faculty of Medicine, University of Maribor, 2000 Maribor, Slovenia
- Correspondence:
| |
Collapse
|
10
|
Autoantibodies Against the Glial Glutamate Transporter GLT1/EAAT2 in Type 1 Diabetes Mellitus. Clues to novel immunological and non-immunological therapies. Pharmacol Res 2022; 177:106130. [DOI: 10.1016/j.phrs.2022.106130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/22/2022]
|
11
|
Abstract
This review focuses on the human pancreatic islet-including its structure, cell composition, development, function, and dysfunction. After providing a historical timeline of key discoveries about human islets over the past century, we describe new research approaches and technologies that are being used to study human islets and how these are providing insight into human islet physiology and pathophysiology. We also describe changes or adaptations in human islets in response to physiologic challenges such as pregnancy, aging, and insulin resistance and discuss islet changes in human diabetes of many forms. We outline current and future interventions being developed to protect, restore, or replace human islets. The review also highlights unresolved questions about human islets and proposes areas where additional research on human islets is needed.
Collapse
Affiliation(s)
- John T Walker
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Diane C Saunders
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Marcela Brissova
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- VA Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
12
|
Ng XW, Chung YH, Piston DW. Intercellular Communication in the Islet of Langerhans in Health and Disease. Compr Physiol 2021; 11:2191-2225. [PMID: 34190340 PMCID: PMC8985231 DOI: 10.1002/cphy.c200026] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Blood glucose homeostasis requires proper function of pancreatic islets, which secrete insulin, glucagon, and somatostatin from the β-, α-, and δ-cells, respectively. Each islet cell type is equipped with intrinsic mechanisms for glucose sensing and secretory actions, but these intrinsic mechanisms alone cannot explain the observed secretory profiles from intact islets. Regulation of secretion involves interconnected mechanisms among and between islet cell types. Islet cells lose their normal functional signatures and secretory behaviors upon dispersal as compared to intact islets and in vivo. In dispersed islet cells, the glucose response of insulin secretion is attenuated from that seen from whole islets, coordinated oscillations in membrane potential and intracellular Ca2+ activity, as well as the two-phase insulin secretion profile, are missing, and glucagon secretion displays higher basal secretion profile and a reverse glucose-dependent response from that of intact islets. These observations highlight the critical roles of intercellular communication within the pancreatic islet, and how these communication pathways are crucial for proper hormonal and nonhormonal secretion and glucose homeostasis. Further, misregulated secretions of islet secretory products that arise from defective intercellular islet communication are implicated in diabetes. Intercellular communication within the islet environment comprises multiple mechanisms, including electrical synapses from gap junctional coupling, paracrine interactions among neighboring cells, and direct cell-to-cell contacts in the form of juxtacrine signaling. In this article, we describe the various mechanisms that contribute to proper islet function for each islet cell type and how intercellular islet communications are coordinated among the same and different islet cell types. © 2021 American Physiological Society. Compr Physiol 11:2191-2225, 2021.
Collapse
Affiliation(s)
- Xue W Ng
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - Yong H Chung
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| | - David W Piston
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri, USA
| |
Collapse
|
13
|
Pietrancosta N, Djibo M, Daumas S, El Mestikawy S, Erickson JD. Molecular, Structural, Functional, and Pharmacological Sites for Vesicular Glutamate Transporter Regulation. Mol Neurobiol 2020; 57:3118-3142. [PMID: 32474835 PMCID: PMC7261050 DOI: 10.1007/s12035-020-01912-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/30/2020] [Indexed: 12/11/2022]
Abstract
Vesicular glutamate transporters (VGLUTs) control quantal size of glutamatergic transmission and have been the center of numerous studies over the past two decades. VGLUTs contain two independent transport modes that facilitate glutamate packaging into synaptic vesicles and phosphate (Pi) ion transport into the synaptic terminal. While a transmembrane proton electrical gradient established by a vacuolar-type ATPase powers vesicular glutamate transport, recent studies indicate that binding sites and flux properties for chloride, potassium, and protons within VGLUTs themselves regulate VGLUT activity as well. These intrinsic ionic binding and flux properties of VGLUTs can therefore be modulated by neurophysiological conditions to affect levels of glutamate available for release from synapses. Despite their extraordinary importance, specific and high-affinity pharmacological compounds that interact with these sites and regulate VGLUT function, distinguish between the various modes of transport, and the different isoforms themselves, are lacking. In this review, we provide an overview of the physiologic sites for VGLUT regulation that could modulate glutamate release in an over-active synapse or in a disease state.
Collapse
Affiliation(s)
- Nicolas Pietrancosta
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Laboratoire des Biomolécules, Sorbonne Université, CNRS, ENS, LBM, 75005, Paris, France.
| | - Mahamadou Djibo
- Sorbonne Paris Cité, Université Paris Descartes, LCBPT, UMR 8601, 75006, Paris, France
| | - Stephanie Daumas
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France
| | - Salah El Mestikawy
- Neuroscience Paris Seine - Institut de Biologie Paris Seine (NPS - IBPS) INSERM, CNRS, Sorbonne Université, Paris, France. .,Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 boulevard Lasalle, Verdun, Montreal, QC, Canada.
| | - Jeffrey D Erickson
- Neuroscience Center, Louisiana State University, New Orleans, LA, 70112, USA. .,Department of Pharmacology, Louisiana State University, New Orleans, LA, 70112, USA.
| |
Collapse
|
14
|
Rodriguez-Diaz R, Tamayo A, Hara M, Caicedo A. The Local Paracrine Actions of the Pancreatic α-Cell. Diabetes 2020; 69:550-558. [PMID: 31882565 PMCID: PMC7085245 DOI: 10.2337/dbi19-0002] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Secretion of glucagon from the pancreatic α-cells is conventionally seen as the first and most important defense against hypoglycemia. Recent findings, however, show that α-cell signals stimulate insulin secretion from the neighboring β-cell. This article focuses on these seemingly counterintuitive local actions of α-cells and describes how they impact islet biology and glucose metabolism. It is mostly based on studies published in the last decade on the physiology of α-cells in human islets and incorporates results from rodents where appropriate. As this and the accompanying articles show, the emerging picture of α-cell function is one of increased complexity that needs to be considered when developing new therapies aimed at promoting islet function in the context of diabetes.
Collapse
Affiliation(s)
- Rayner Rodriguez-Diaz
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Alejandro Tamayo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
| | - Manami Hara
- Department of Medicine, University of Chicago, Chicago, IL
| | - Alejandro Caicedo
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL
- Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, FL
- Program in Neuroscience, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
15
|
Abstract
Glucagon and its partner insulin are dually linked in both their secretion from islet cells and their action in the liver. Glucagon signaling increases hepatic glucose output, and hyperglucagonemia is partly responsible for the hyperglycemia in diabetes, making glucagon an attractive target for therapeutic intervention. Interrupting glucagon signaling lowers blood glucose but also results in hyperglucagonemia and α-cell hyperplasia. Investigation of the mechanism for α-cell proliferation led to the description of a conserved liver-α-cell axis where glucagon is a critical regulator of amino acid homeostasis. In return, amino acids regulate α-cell function and proliferation. New evidence suggests that dysfunction of the axis in humans may result in the hyperglucagonemia observed in diabetes. This discussion outlines important but often overlooked roles for glucagon that extend beyond glycemia and supports a new role for α-cells as amino acid sensors.
Collapse
Affiliation(s)
- E Danielle Dean
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
16
|
Abarkan M, Gaitan J, Lebreton F, Perrier R, Jaffredo M, Mulle C, Magnan C, Raoux M, Lang J. The glutamate receptor GluK2 contributes to the regulation of glucose homeostasis and its deterioration during aging. Mol Metab 2019; 30:152-160. [PMID: 31767166 PMCID: PMC6807305 DOI: 10.1016/j.molmet.2019.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/04/2019] [Accepted: 09/27/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE Islets secrete neurotransmitters including glutamate which participate in fine regulation of islet function. The excitatory ionotropic glutamate receptor GluK2 of the kainate receptor family is widely expressed in brain and also found in islets, mainly in α and γ cells. α cells co-release glucagon and glutamate and the latter increases glucagon release via ionotropic glutamate receptors. However, neither the precise nature of the ionotropic glutamate receptor involved nor its role in glucose homeostasis is known. As isoform specific pharmacology is not available, we investigated this question in constitutive GluK2 knock-out mice (GluK2-/-) using adult and middle-aged animals to also gain insight in a potential role during aging. METHODS We compared wild-type GluK2+/+ and knock-out GluK2-/- mice using adult (14-20 weeks) and middle-aged animals (40-52 weeks). Glucose (oral OGTT and intraperitoneal IPGTT) and insulin tolerance as well as pyruvate challenge tests were performed according to standard procedures. Parasympathetic activity, which stimulates hormones secretion, was measured by electrophysiology in vivo. Isolated islets were used in vitro to determine islet β-cell electrical activity on multi-electrode arrays and dynamic secretion of insulin as well as glucagon was determined by ELISA. RESULTS Adult GluK2-/- mice exhibit an improved glucose tolerance (OGTT and IPGTT), and this was also apparent in middle-aged mice, whereas the outcome of pyruvate challenge was slightly improved only in middle-aged GluK2-/- mice. Similarly, insulin sensitivity was markedly enhanced in middle-aged GluK2-/- animals. Basal and glucose-induced insulin secretion in vivo was slightly lower in GluK2-/- mice, whereas fasting glucagonemia was strongly reduced. In vivo recordings of parasympathetic activity showed an increase in basal activity in GluK2-/- mice which represents most likely an adaptive mechanism to counteract hypoglucagonemia rather than altered neuronal mechanism. In vitro recording demonstrated an improvement of glucose-induced electrical activity of β-cells in islets obtained from GluK2-/- mice at both ages. Finally, glucose-induced insulin secretion in vitro was increased in GluK2-/- islets, whereas glucagon secretion at 2 mmol/l of glucose was considerably reduced. CONCLUSIONS These observations indicate a general role for kainate receptors in glucose homeostasis and specifically suggest a negative effect of GluK2 on glucose homeostasis and preservation of islet function during aging. Our observations raise the possibility that blockade of GluK2 may provide benefits in glucose homeostasis especially during aging.
Collapse
Affiliation(s)
- Myriam Abarkan
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Julien Gaitan
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Fanny Lebreton
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Romain Perrier
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Manon Jaffredo
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Christophe Mulle
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Université de Bordeaux, Bordeaux, France
| | - Christophe Magnan
- Unité de Biologie Fonctionnelle et Adaptative, UMR 8251, CNRS, Université de Paris, Paris, France
| | - Matthieu Raoux
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France
| | - Jochen Lang
- Chimie et Biologie des Membranes et Nano-objets, UMR CNRS 5248, Université de Bordeaux, Pessac, France.
| |
Collapse
|
17
|
Anton Dib Saleh M, Sousa Dos Santos L, Antonio Berto D, Borges Amorim A, Lívio Panhoza Tse M, Eliodoro Costa V. IRMS as a tool to obtain the carbon turnover (δ 13 C) in organs of weaned piglets fed glutamic acid and nucleotides. J Anim Physiol Anim Nutr (Berl) 2019; 103:906-914. [PMID: 30924561 DOI: 10.1111/jpn.13089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
Early weaning of piglets causes stress characterized by a decrease in feed intake followed by a decline in growth rates; thus, a fast recovery represents an essential step for proper growth of these animals. Considering that IRMS is a potential tool for non-destructive sampling and the fact that it provides time-integrated estimate of assimilated and not just ingested nutrients turned possible its application to evaluate the effects of dietary nucleotides and glutamate on carbon turnover (δ13 C) in organs of weanling piglets. At day 0, three piglets were slaughtered (prior to diet switch), the remaining eighty-four piglets weaned at 21-day-old were randomly assigned in a complete block design with a 2 × 2 factorial arrangement of treatments (two Nu levels: 0 and 0.1% and two Glu levels: 0 and 1%), being three piglets per treatment slaughtered on trial days 3, 6, 9, 14, 21, 35 and 49. The samples were analysed by IRMS and adjusted to first-order equation by a non-linear regression analysis using NLIN of SAS, in order to establish exponential graphics. After that, the turnover data were submitted to analysis of variance using GLM of SAS. The turnover value (t95% ) verified for spleen was faster (p < 0.05) when glutamate was supplemented in diets. For pancreas and liver, the turnover rates were faster (p < 0.05) for the mixture of additives. However, for renal tissue, the turnover rate (t95% ) was greater (p < 0.05) for the free additive diet. The results obtained suggest that the mixture of additives was more efficient to develop the digestive tract at post-weaning phase, taking into account the functional importance of pancreas and liver for nutrients' digestion and processing.
Collapse
Affiliation(s)
- Mayra Anton Dib Saleh
- Department of Animal Production, UNESP - São Paulo State University, Botucatu, Brazil
| | - Luan Sousa Dos Santos
- Department of Animal Production, UNESP - São Paulo State University, Botucatu, Brazil
| | - Dirlei Antonio Berto
- Department of Animal Production, UNESP - São Paulo State University, Botucatu, Brazil
| | - Alessandro Borges Amorim
- Institute of Agricultural Sciences and Technologies, Federal University of Mato Grosso, Rondonópolis, Brazil
| | | | - Vladimir Eliodoro Costa
- Institute of Biosciences, Environmental Stable Isotopes Center, UNESP - São Paulo State University, Botucatu, Brazil
| |
Collapse
|
18
|
Asadi F, Dhanvantari S. Plasticity in the Glucagon Interactome Reveals Novel Proteins That Regulate Glucagon Secretion in α-TC1-6 Cells. Front Endocrinol (Lausanne) 2019; 9:792. [PMID: 30713523 PMCID: PMC6346685 DOI: 10.3389/fendo.2018.00792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 12/17/2018] [Indexed: 12/27/2022] Open
Abstract
Glucagon is stored within the secretory granules of pancreatic alpha cells until stimuli trigger its release. The alpha cell secretory responses to the stimuli vary widely, possibly due to differences in experimental models or microenvironmental conditions. We hypothesized that the response of the alpha cell to various stimuli could be due to plasticity in the network of proteins that interact with glucagon within alpha cell secretory granules. We used tagged glucagon with Fc to pull out glucagon from the enriched preparation of secretory granules in α-TC1-6 cells. Isolation of secretory granules was validated by immunoisolation with Fc-glucagon and immunoblotting for organelle-specific proteins. Isolated enriched secretory granules were then used for affinity purification with Fc-glucagon followed by liquid chromatography/tandem mass spectrometry to identify secretory granule proteins that interact with glucagon. Proteomic analyses revealed a network of proteins containing glucose regulated protein 78 KDa (GRP78) and histone H4. The interaction between glucagon and the ER stress protein GRP78 and histone H4 was confirmed through co-immunoprecipitation of secretory granule lysates, and colocalization immunofluorescence confocal microscopy. Composition of the protein networks was altered at different glucose levels (25 vs. 5.5 mM) and in response to the paracrine inhibitors of glucagon secretion, GABA and insulin. siRNA-mediated silencing of a subset of these proteins revealed their involvement in glucagon secretion in α-TC1-6 cells. Therefore, our results show a novel and dynamic glucagon interactome within α-TC1-6 cell secretory granules. We suggest that variations in the alpha cell secretory response to stimuli may be governed by plasticity in the glucagon "interactome."
Collapse
Affiliation(s)
- Farzad Asadi
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Savita Dhanvantari
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Metabolism, Diabetes and Imaging Programs, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
19
|
The UBR-1 ubiquitin ligase regulates glutamate metabolism to generate coordinated motor pattern in Caenorhabditis elegans. PLoS Genet 2018; 14:e1007303. [PMID: 29649217 PMCID: PMC5931689 DOI: 10.1371/journal.pgen.1007303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 05/02/2018] [Accepted: 03/08/2018] [Indexed: 12/14/2022] Open
Abstract
UBR1 is an E3 ubiquitin ligase best known for its ability to target protein degradation by the N-end rule. The physiological functions of UBR family proteins, however, remain not fully understood. We found that the functional loss of C. elegans UBR-1 leads to a specific motor deficit: when adult animals generate reversal movements, A-class motor neurons exhibit synchronized activation, preventing body bending. This motor deficit is rescued by removing GOT-1, a transaminase that converts aspartate to glutamate. Both UBR-1 and GOT-1 are expressed and critically required in premotor interneurons of the reversal motor circuit to regulate the motor pattern. ubr-1 and got-1 mutants exhibit elevated and decreased glutamate level, respectively. These results raise an intriguing possibility that UBR proteins regulate glutamate metabolism, which is critical for neuronal development and signaling. Ubiquitin-mediated protein degradation is central to diverse biological processes. The selection of substrates for degradation is carried out by the E3 ubiquitin ligases, which target specific groups of proteins for ubiquitination. The human genome encodes hundreds of E3 ligases; many exhibit sequence conservation across animal species, including one such ligase called UBR1. Patients carrying mutations in UBR1 exhibit severe systemic defects, but the biology behinds UBR1’s physiological function remains elusive. Here we found that the C. elegans UBR-1 regulates glutamate level. When UBR-1 is defective, C. elegans exhibits increased glutamate; this leads to synchronization of motor neuron activity, hence defective locomotion when animals reach adulthood. UBR1-mediated glutamate metabolism may contribute to the physiological defects of UBR1 mutations.
Collapse
|
20
|
Zemková H, Stojilkovic SS. Neurotransmitter receptors as signaling platforms in anterior pituitary cells. Mol Cell Endocrinol 2018; 463:49-64. [PMID: 28684290 PMCID: PMC5752632 DOI: 10.1016/j.mce.2017.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/29/2017] [Accepted: 07/02/2017] [Indexed: 02/07/2023]
Abstract
The functions of anterior pituitary cells are controlled by two major groups of hypothalamic and intrapituitary ligands: one exclusively acts on G protein-coupled receptors and the other activates both G protein-coupled receptors and ligand-gated receptor channels. The second group of ligands operates as neurotransmitters in neuronal cells and their receptors are termed as neurotransmitter receptors. Most information about pituitary neurotransmitter receptors was obtained from secretory studies, RT-PCR analyses of mRNA expression and immunohistochemical and biochemical analyses, all of which were performed using a mixed population of pituitary cells. However, recent electrophysiological and imaging experiments have characterized γ-aminobutyric acid-, acetylcholine-, and ATP-activated receptors and channels in single pituitary cell types, expanding this picture and revealing surprising differences in their expression between subtypes of secretory cells and between native and immortalized pituitary cells. The main focus of this review is on the electrophysiological and pharmacological properties of these receptors and their roles in calcium signaling and calcium-controlled hormone secretion.
Collapse
Affiliation(s)
- Hana Zemková
- Department of Cellular and Molecular Neuroendocrinology, Institute of Physiology, ASCR, Prague, Czech Republic.
| | - Stanko S Stojilkovic
- Sections on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
21
|
Andersson LE, Shcherbina L, Al-Majdoub M, Vishnu N, Arroyo CB, Aste Carrara J, Wollheim CB, Fex M, Mulder H, Wierup N, Spégel P. Glutamine-Elicited Secretion of Glucagon-Like Peptide 1 Is Governed by an Activated Glutamate Dehydrogenase. Diabetes 2018; 67:372-384. [PMID: 29229616 DOI: 10.2337/db16-1441] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 12/07/2017] [Indexed: 11/13/2022]
Abstract
Glucagon-like peptide 1 (GLP-1), secreted from intestinal L cells, glucose dependently stimulates insulin secretion from β-cells. This glucose dependence prevents hypoglycemia, rendering GLP-1 analogs a useful and safe treatment modality in type 2 diabetes. Although the amino acid glutamine is a potent elicitor of GLP-1 secretion, the responsible mechanism remains unclear. We investigated how GLP-1 secretion is metabolically coupled in L cells (GLUTag) and in vivo in mice using the insulin-secreting cell line INS-1 832/13 as reference. A membrane-permeable glutamate analog (dimethylglutamate [DMG]), acting downstream of electrogenic transporters, elicited similar alterations in metabolism as glutamine in both cell lines. Both DMG and glutamine alone elicited GLP-1 secretion in GLUTag cells and in vivo, whereas activation of glutamate dehydrogenase (GDH) was required to stimulate insulin secretion from INS-1 832/13 cells. Pharmacological inhibition in vivo of GDH blocked secretion of GLP-1 in response to DMG. In conclusion, our results suggest that nonelectrogenic nutrient uptake and metabolism play an important role in L cell stimulus-secretion coupling. Metabolism of glutamine and related analogs by GDH in the L cell may explain why GLP-1 secretion, but not that of insulin, is activated by these secretagogues in vivo.
Collapse
Affiliation(s)
- Lotta E Andersson
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Liliya Shcherbina
- Neuroendocrine Cell Biology, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Mahmoud Al-Majdoub
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Neelanjan Vishnu
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | | | - Jonathan Aste Carrara
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| | - Claes B Wollheim
- Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
- Department of Cell Physiology and Metabolism, University Medical Centre, Geneva, Switzerland
| | - Malin Fex
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Hindrik Mulder
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Nils Wierup
- Neuroendocrine Cell Biology, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
| | - Peter Spégel
- Unit of Molecular Metabolism, Department of Clinical Sciences, Lund University Diabetes Centre, Clinical Research Centre, Skåne University Hospital, Malmö, Sweden
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University, Lund, Sweden
| |
Collapse
|
22
|
Moriyama S, Iharada M, Omote H, Moriyama Y, Hiasa M. Function and expression of a splicing variant of vesicular glutamate transporter 1. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:931-940. [PMID: 28188742 DOI: 10.1016/j.bbamem.2017.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/30/2017] [Accepted: 02/05/2017] [Indexed: 10/20/2022]
Abstract
Vesicular glutamate transporter (VGLUT) is an active transporter responsible for vesicular storage of glutamate in synaptic vesicles and plays an essential role in glutamatergic neurotransmission. VGLUT consists of three isoforms, VGLUT1, VGLUT2, and VGLUT3. The VGLUT1 variant, VGLUT1v, with an additional 75-base pair sequence derived from a second intron between exons 2 and 3, which corresponds to 25 amino acid residues in the 1st loop of VGLUT1, is the only splicing variant among VGLUTs, although whether VGLUT1v protein is actually translated at the protein level remains unknown. In the present study, VGLUT1v was expressed in insect cells, solubilized, purified to near homogeneity, and its transport activity was examined. Proteoliposomes containing purified VGLUT1v were shown to accumulate glutamate upon imposition of an inside-positive membrane potential (Δψ). The Δψ-driven glutamate uptake activity requires Cl- and its pharmacological profile and kinetics are comparable to those of other VGLUTs. The retinal membrane contained two VGLUT1 moieties with apparent molecular masses of 65 and 57kDa. VGLUT1v-specific antibodies against an inserted 25-amino acid residue sequence identified a 65-kDa immunoreactive polypeptide. Immunohistochemical analysis indicated that VGLUT1v immunoreactivity is present in photoreceptor cells and is associated with synaptic vesicles. VGLUT1v immunoreactivity is also present in pinealocytes, but not in other areas, including the brain. These results indicated that VGLUT1v exists in a functional state in rat photosensitive cells and is involved in glutamatergic chemical transmission.
Collapse
Affiliation(s)
- Satomi Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Masafumi Iharada
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan
| | - Yoshinori Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan.
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan.
| |
Collapse
|
23
|
Takeuchi T, Harada Y, Moriyama S, Furuta K, Tanaka S, Miyaji T, Omote H, Moriyama Y, Hiasa M. Vesicular Polyamine Transporter Mediates Vesicular Storage and Release of Polyamine from Mast Cells. J Biol Chem 2017; 292:3909-3918. [PMID: 28082679 DOI: 10.1074/jbc.m116.756197] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/04/2017] [Indexed: 01/12/2023] Open
Abstract
Mast cells are secretory cells that play an important role in host defense by discharging various intragranular contents, such as histamine and serotonin, upon stimulation of Fc receptors. The granules also contain spermine and spermidine, which can act as modulators of mast cell function, although the mechanism underlying vesicular storage remains unknown. Vesicular polyamine transporter (VPAT), the fourth member of the SLC18 transporter family, is an active transporter responsible for vesicular storage of spermine and spermidine in neurons. In the present study, we investigated whether VPAT functions in mast cells. RT-PCR and Western blotting indicated VPAT expression in murine bone marrow-derived mast cells (BMMCs). Immunohistochemical analysis indicated that VPAT is colocalized with VAMP3 but not with histamine, serotonin, cathepsin D, VAMP2, or VAMP7. Membrane vesicles from BMMCs accumulated spermidine upon the addition of ATP in a reserpine- and bafilomycin A1-sensitive manner. BMMCs secreted spermine and spermidine upon the addition of either antigen or A23187 in the presence of Ca2+, and the antigen-mediated release, which was shown to be temperature-dependent and sensitive to bafilomycin A1 and tetanus toxin, was significantly suppressed by VPAT gene RNA interference. Under these conditions, expression of vesicular monoamine transporter 2 was unaffected, but antigen-dependent histamine release was significantly suppressed, which was recovered by the addition of 1 mm spermine. These results strongly suggest that VPAT is expressed and is responsible for vesicular storage of spermine and spermidine in novel secretory granules that differ from histamine- and serotonin-containing granules and is involved in vesicular release of these polyamines from mast cells.
Collapse
Affiliation(s)
- Tomoya Takeuchi
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530
| | - Yuika Harada
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530
| | - Satomi Moriyama
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530
| | - Kazuyuki Furuta
- the Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8530, and
| | - Satoshi Tanaka
- the Department of Immunobiology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8530, and
| | - Takaaki Miyaji
- the Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Hiroshi Omote
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530
| | - Yoshinori Moriyama
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530,
| | - Miki Hiasa
- From the Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530,
| |
Collapse
|
24
|
Moriyama S, Hiasa M. Expression of Vesicular Nucleotide Transporter in the Mouse Retina. Biol Pharm Bull 2017; 39:564-9. [PMID: 27040629 DOI: 10.1248/bpb.b15-00872] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vesicular nucleotide transporter (VNUT) is a membrane protein that is responsible for vesicular storage and subsequent vesicular release of nucleotides, such as ATP, and plays an essential role in purinergic chemical transmission. In the present study, we investigated whether VNUT is present in the rodent retina to define the site(s) of vesicular ATP release. In the mouse retina, reverse transcription polymerase chain reaction (RT-PCR) and immunological analyses using specific anti-VNUT antibodies indicated that VNUT is expressed as a polypeptide with an apparent molecular mass of 59 kDa. VNUT is widely distributed throughout the inner and outer retinal layers, particularly in the outer segment of photoreceptors, outer plexiform layer, inner plexiform layer, and ganglion cell layer. VNUT is colocalized with vesicular glutamate transporter 1 and synaptophysin in photoreceptor cells, while it is colocalized with vesicular γ-aminobutyric acid (GABA) transporter in amacrine cells and bipolar cells. VNUT is also present in astrocytes and Müller cells. The retina from VNUT knockout (VNUT(-/-)) mice showed the loss of VNUT immunoreactivity. The retinal membrane fraction took up radiolabeled ATP in diisothiocyanate stilbene disulfonic acid (DIDS)-, an inhibitor of VNUT, and bafilomycin A1-, a vacuolar adenosine triphosphatase (ATPase) inhibitor, in a sensitive manner, while membranes from VNUT(-/-) mice showed the loss of DIDS-sensitive ATP uptake. Taken together, these results indicate that functional VNUT is expressed in the rodent retina and suggest that ATP is released from photoreceptor cells, bipolar cells, amacrine cells, and astrocytes as well as Müller cells to initiate purinergic chemical transmission.
Collapse
Affiliation(s)
- Satomi Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences
| | | |
Collapse
|
25
|
Otter S, Lammert E. Exciting Times for Pancreatic Islets: Glutamate Signaling in Endocrine Cells. Trends Endocrinol Metab 2016; 27:177-188. [PMID: 26740469 DOI: 10.1016/j.tem.2015.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 01/14/2023]
Abstract
Glutamate represents a key excitatory neurotransmitter in the central nervous system, and also modulates the function and viability of endocrine cells in pancreatic islets. In insulin-secreting beta cells, glutamate acts as an intracellular messenger, and its transport into secretory granules promotes glucose- and incretin-stimulated insulin secretion. Mitochondrial degradation of glutamate also contributes to insulin release when glutamate dehydrogenase is allosterically activated. It also signals extracellularly via glutamate receptors (AMPA and NMDA receptors) to modulate glucagon, insulin and somatostatin secretion, and islet cell survival. Its degradation products, GABA and γ-hydroxybutyrate, are released and also influence islet cell behavior. Thus, islet glutamate receptors, such as the NMDA receptors, might serve as possible drug targets to develop new medications for adjunct treatment of diabetes.
Collapse
Affiliation(s)
- Silke Otter
- Institute of Metabolic Physiology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, and German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany
| | - Eckhard Lammert
- Institute of Metabolic Physiology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; Institute for Beta Cell Biology, German Diabetes Center (DDZ), Leibniz Center for Diabetes Research, and German Center for Diabetes Research (DZD e.V.), Düsseldorf, Germany.
| |
Collapse
|
26
|
|
27
|
Omote H, Miyaji T, Hiasa M, Juge N, Moriyama Y. Structure, Function, and Drug Interactions of Neurotransmitter Transporters in the Postgenomic Era. Annu Rev Pharmacol Toxicol 2015; 56:385-402. [PMID: 26514205 DOI: 10.1146/annurev-pharmtox-010814-124816] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Vesicular neurotransmitter transporters are responsible for the accumulation of neurotransmitters in secretory vesicles and play essential roles in chemical transmission. The SLC17 family contributes to sequestration of anionic neurotransmitters such as glutamate, aspartate, and nucleotides. Identification and subsequent cellular and molecular biological studies of SLC17 transporters unveiled the principles underlying the actions of these transporters. Recent progress in reconstitution methods in combination with postgenomic approaches has advanced studies on neurotransmitter transporters. This review summarizes the molecular properties of SLC17-type transporters and recent findings regarding the novel SLC18 transporter.
Collapse
Affiliation(s)
- Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; ,
| | - Takaaki Miyaji
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; ,
| | - Narinobu Juge
- Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| | - Yoshinori Moriyama
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, Japan; , .,Advanced Science Research Center, Okayama University, Okayama 700-8530, Japan
| |
Collapse
|
28
|
Arrojo e Drigo R, Ali Y, Diez J, Srinivasan DK, Berggren PO, Boehm BO. New insights into the architecture of the islet of Langerhans: a focused cross-species assessment. Diabetologia 2015. [PMID: 26215305 DOI: 10.1007/s00125-015-3699-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The human genome project and its search for factors underlying human diseases has fostered a major human research effort. Therefore, unsurprisingly, in recent years we have observed an increasing number of studies on human islet cells, including disease approaches focusing on type 1 and type 2 diabetes. Yet, the field of islet and diabetes research relies on the legacy of rodent-based investigations, which have proven difficult to translate to humans, particularly in type 1 diabetes. Whole islet physiology and pathology may differ between rodents and humans, and thus a comprehensive cross-species as well as species-specific view on islet research is much needed. In this review we summarise the current knowledge of interspecies islet cytoarchitecture, and discuss its potential impact on islet function and future perspectives in islet pathophysiology research.
Collapse
Affiliation(s)
- Rafael Arrojo e Drigo
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Yusuf Ali
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Juan Diez
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Dinesh Kumar Srinivasan
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore
| | - Per-Olof Berggren
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska University Hospital L1, Karolinska Institutet, SE-171 76, Stockholm, Sweden.
| | - Bernhard O Boehm
- Lee Kong Chian School of Medicine, Nanyang Technological University, 50 Nanyang Drive, Research Techno Plaza, Level 4, 637 553, Singapore, Singapore.
- Imperial College London, London, UK.
- Department of Internal Medicine 1, Ulm University Medical Centre, Ulm, Germany.
| |
Collapse
|
29
|
Di Cairano ES, Moretti S, Marciani P, Sacchi VF, Castagna M, Davalli A, Folli F, Perego C. Neurotransmitters and Neuropeptides: New Players in the Control of Islet of Langerhans' Cell Mass and Function. J Cell Physiol 2015; 231:756-67. [PMID: 26332080 DOI: 10.1002/jcp.25176] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/27/2015] [Indexed: 12/19/2022]
Abstract
Islets of Langerhans control whole body glucose homeostasis, as they respond, releasing hormones, to changes in nutrient concentrations in the blood stream. The regulation of hormone secretion has been the focus of attention for a long time because it is related to many metabolic disorders, including diabetes mellitus. Endocrine cells of the islet use a sophisticate system of endocrine, paracrine and autocrine signals to synchronize their activities. These signals provide a fast and accurate control not only for hormone release but also for cell differentiation and survival, key aspects in islet physiology and pathology. Among the different categories of paracrine/autocrine signals, this review highlights the role of neurotransmitters and neuropeptides. In a manner similar to neurons, endocrine cells synthesize, accumulate, release neurotransmitters in the islet milieu, and possess receptors able to decode these signals. In this review, we provide a comprehensive description of neurotransmitter/neuropetide signaling pathways present within the islet. Then, we focus on evidence supporting the concept that neurotransmitters/neuropeptides and their receptors are interesting new targets to preserve β-cell function and mass. A greater understanding of how this network of signals works in physiological and pathological conditions would advance our knowledge of islet biology and physiology and uncover potentially new areas of pharmacological intervention. J. Cell. Physiol. 231: 756-767, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Eliana S Di Cairano
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Stefania Moretti
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Paola Marciani
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Vellea Franca Sacchi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Michela Castagna
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| | - Alberto Davalli
- Department of Internal Medicine, Diabetes and Endocrinology Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Franco Folli
- Division of Diabetes, Department of Medicine, University of Texas Health Science Center at San Antonio, Texas.,Department of Internal Medicine, Obesity and Comorbidities Research Center (OCRC), University of Campinas, UNICAMP, Campinas, Sao Paulo State, Brazil
| | - Carla Perego
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Laboratory of Molecular and Cellular Physiology, Universit, à, degli Studi di Milano, Milan, Italy
| |
Collapse
|
30
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
31
|
Yin W, Sun Z, Mendenhall JM, Walker DM, Riha PD, Bezner KS, Gore AC. Expression of Vesicular Glutamate Transporter 2 (vGluT2) on Large Dense-Core Vesicles within GnRH Neuroterminals of Aging Female Rats. PLoS One 2015; 10:e0129633. [PMID: 26053743 PMCID: PMC4459826 DOI: 10.1371/journal.pone.0129633] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/10/2015] [Indexed: 11/20/2022] Open
Abstract
The pulsatile release of GnRH is crucial for normal reproductive physiology across the life cycle, a process that is regulated by hypothalamic neurotransmitters. GnRH terminals co-express the vesicular glutamate transporter 2 (vGluT2) as a marker of a glutamatergic phenotype. The current study sought to elucidate the relationship between glutamate and GnRH nerve terminals in the median eminence—the site of GnRH release into the portal capillary vasculature. We also determined whether this co-expression may change during reproductive senescence, and if steroid hormones, which affect responsiveness of GnRH neurons to glutamate, may alter the co-expression pattern. Female Sprague-Dawley rats were ovariectomized at young adult, middle-aged and old ages (~4, 11, and 22 months, respectively) and treated four weeks later with sequential vehicle + vehicle (VEH + VEH), estradiol + vehicle (E2 + VEH), or estradiol + progesterone (E2+P4). Rats were perfused 24 hours after the second hormone treatment. Confocal microscopy was used to determine colocalization of GnRH and vGluT2 immunofluorescence in the median eminence. Post-embedding immunogold labeling of GnRH and vGluT2, and a serial electron microscopy (EM) technique were used to determine the cellular interaction between GnRH terminals and glutamate signaling. Confocal analysis showed that GnRH and vGluT2 immunofluorescent puncta were extensively colocalized in the median eminence and that their density declined with age but was unaffected by short-term hormone treatment. EM results showed that vGluT2 immunoreactivity was extensively associated with large dense-core vesicles, suggesting a unique glutamatergic signaling pathway in GnRH terminals. Our results provide novel subcellular information about the intimate relationship between GnRH terminals and glutamate in the median eminence.
Collapse
Affiliation(s)
- Weiling Yin
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Zengrong Sun
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - John M. Mendenhall
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Deena M. Walker
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
| | - Penny D. Riha
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Kelsey S. Bezner
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
| | - Andrea C. Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, University of Texas at Austin, Austin, Texas, United States of America
- Institute for Neuroscience, University of Texas at Austin, Austin, Texas, United States of America
- Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Kanaani J, Cianciaruso C, Phelps EA, Pasquier M, Brioudes E, Billestrup N, Baekkeskov S. Compartmentalization of GABA synthesis by GAD67 differs between pancreatic beta cells and neurons. PLoS One 2015; 10:e0117130. [PMID: 25647668 PMCID: PMC4315522 DOI: 10.1371/journal.pone.0117130] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 12/19/2014] [Indexed: 11/22/2022] Open
Abstract
The inhibitory neurotransmitter GABA is synthesized by the enzyme glutamic acid decarboxylase (GAD) in neurons and in pancreatic β-cells in islets of Langerhans where it functions as a paracrine and autocrine signaling molecule regulating the function of islet endocrine cells. The localization of the two non-allelic isoforms GAD65 and GAD67 to vesicular membranes is important for rapid delivery and accumulation of GABA for regulated secretion. While the membrane anchoring and trafficking of GAD65 are mediated by intrinsic hydrophobic modifications, GAD67 remains hydrophilic, and yet is targeted to vesicular membrane pathways and synaptic clusters in neurons by both a GAD65-dependent and a distinct GAD65-independent mechanism. Herein we have investigated the membrane association and targeting of GAD67 and GAD65 in monolayer cultures of primary rat, human, and mouse islets and in insulinoma cells. GAD65 is primarily detected in Golgi membranes and in peripheral vesicles distinct from insulin vesicles in β-cells. In the absence of GAD65, GAD67 is in contrast primarily cytosolic in β-cells; its co-expression with GAD65 is necessary for targeting to Golgi membranes and vesicular compartments. Thus, the GAD65-independent mechanism for targeting of GAD67 to synaptic vesicles in neurons is not functional in islet β-cells. Therefore, only GAD65:GAD65 homodimers and GAD67:GAD65 heterodimers, but not the GAD67:GAD67 homodimer gain access to vesicular compartments in β-cells to facilitate rapid accumulation of newly synthesized GABA for regulated secretion and fine tuning of GABA-signaling in islets of Langerhans.
Collapse
Affiliation(s)
- Jamil Kanaani
- Departments of Medicine and Microbiology/Immunology, Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
| | - Chiara Cianciaruso
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Edward A. Phelps
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Miriella Pasquier
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Estelle Brioudes
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Nils Billestrup
- Institute of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steinunn Baekkeskov
- Departments of Medicine and Microbiology/Immunology, Diabetes Center, University of California San Francisco, San Francisco, California, United States of America
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
33
|
Bessho M, Murase-Mishiba Y, Imagawa A, Terasaki J, Hanafusa T. Possible contribution of taurine to distorted glucagon secretion in intra-islet insulin deficiency: a metabolome analysis using a novel α-cell model of insulin-deficient diabetes. PLoS One 2014; 9:e113254. [PMID: 25393115 PMCID: PMC4231115 DOI: 10.1371/journal.pone.0113254] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 10/24/2014] [Indexed: 01/30/2023] Open
Abstract
Glycemic instability is a serious problem in patients with insulin-deficient diabetes, and it may be due in part to abnormal endogenous glucagon secretion. However, the intracellular metabolic mechanism(s) involved in the aberrant glucagon response under the condition of insulin deficiency has not yet been elucidated. To investigate the metabolic traits that underlie the distortion of glucagon secretion under insulin deficient conditions, we generated an αTC1-6 cell line with stable knockdown of the insulin receptor (IRKD), i.e., an in vitro α-cell model for insulin-deficient diabetes, which exhibits an abnormal glucagon response to glucose. A comprehensive metabolomic analysis of the IRKD αTC1-6 cells (IRKD cells) revealed some candidate metabolites whose levels differed markedly compared to those in control αTC1-6 cells, but also which could affect the glucagon release in IRKD cells. Of these candidates, taurine was remarkably increased in the IRKD cells and was identified as a stimulator of glucagon in αTC1-6 cells. Taurine also paradoxically exaggerated the glucagon secretion at a high glucose concentration in IRKD cells and islets with IRKD. These results indicate that the metabolic alterations induced by IRKD in α-cells, especially the increase of taurine, may lead to the distorted glucagon response in IRKD cells, suggesting the importance of taurine in the paradoxical glucagon response and the resultant glucose instability in insulin-deficient diabetes.
Collapse
Affiliation(s)
- Megumi Bessho
- Department of Internal Medicine (I), Osaka Medical College, Osaka, Japan
| | | | - Akihisa Imagawa
- Department of Internal Medicine (I), Osaka Medical College, Osaka, Japan
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Jungo Terasaki
- Department of Internal Medicine (I), Osaka Medical College, Osaka, Japan
| | - Toshiaki Hanafusa
- Department of Internal Medicine (I), Osaka Medical College, Osaka, Japan
| |
Collapse
|
34
|
Identification of a mammalian vesicular polyamine transporter. Sci Rep 2014; 4:6836. [PMID: 25355561 PMCID: PMC4213795 DOI: 10.1038/srep06836] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/10/2014] [Indexed: 01/11/2023] Open
Abstract
Spermine and spermidine act as neuromodulators upon binding to the extracellular site(s) of various ionotropic receptors, such as N-methyl-d-aspartate receptors. To gain access to the receptors, polyamines synthesized in neurons and astrocytes are stored in secretory vesicles and released upon depolarization. Although vesicular storage is mediated in an ATP-dependent, reserpine-sensitive fashion, the transporter responsible for this process remains unknown. SLC18B1 is the fourth member of the SLC18 transporter family, which includes vesicular monoamine transporters and vesicular acetylcholine transporter. Proteoliposomes containing purified human SLC18B1 protein actively transport spermine and spermidine by exchange of H(+). SLC18B1 protein is predominantly expressed in the hippocampus and is associated with vesicles in astrocytes. SLC18B1 gene knockdown decreased both SLC18B1 protein and spermine/spermidine contents in astrocytes. These results indicated that SLC18B1 encodes a vesicular polyamine transporter (VPAT).
Collapse
|
35
|
Sakamoto S, Miyaji T, Hiasa M, Ichikawa R, Uematsu A, Iwatsuki K, Shibata A, Uneyama H, Takayanagi R, Yamamoto A, Omote H, Nomura M, Moriyama Y. Impairment of vesicular ATP release affects glucose metabolism and increases insulin sensitivity. Sci Rep 2014; 4:6689. [PMID: 25331291 PMCID: PMC4204045 DOI: 10.1038/srep06689] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/17/2014] [Indexed: 12/04/2022] Open
Abstract
Neuroendocrine cells store ATP in secretory granules and release it along with hormones that may trigger a variety of cellular responses in a process called purinergic chemical transmission. Although the vesicular nucleotide transporter (VNUT) has been shown to be involved in vesicular storage and release of ATP, its physiological relevance in vivo is far less well understood. In Vnut knockout (Vnut(-/-)) mice, we found that the loss of functional VNUT in adrenal chromaffin granules and insulin granules in the islets of Langerhans led to several significant effects. Vesicular ATP accumulation and depolarization-dependent ATP release were absent in the chromaffin granules of Vnut(-/-) mice. Glucose-responsive ATP release was also absent in pancreatic β-cells in Vnut(-/-) mice, while glucose-responsive insulin secretion was enhanced to a greater extent than that in wild-type tissue. Vnut(-/-) mice exhibited improved glucose tolerance and low blood glucose upon fasting due to increased insulin sensitivity. These results demonstrated an essential role of VNUT in vesicular storage and release of ATP in neuroendocrine cells in vivo and suggest that vesicular ATP and/or its degradation products act as feedback regulators in catecholamine and insulin secretion, thereby regulating blood glucose homeostasis.
Collapse
Affiliation(s)
- Shohei Sakamoto
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Takaaki Miyaji
- Advanced Research Science Center, Okayama University, Okayama 700-8530, JAPAN
| | - Miki Hiasa
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| | - Reiko Ichikawa
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Akira Uematsu
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Ken Iwatsuki
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Atsushi Shibata
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Hisayuki Uneyama
- Institute for Innovation, Ajinomoto Co., Inc. Kawasaki 210-5893, JAPAN
| | - Ryoichi Takayanagi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Akitsugu Yamamoto
- Faculty of Bioscience, Nagahama Institute of Bio-science and Technology, Nagahama 526-0829, JAPAN
| | - Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| | - Masatoshi Nomura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, JAPAN
| | - Yoshinori Moriyama
- Advanced Research Science Center, Okayama University, Okayama 700-8530, JAPAN
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8530, JAPAN
| |
Collapse
|
36
|
Expression of vesicular glutamate transporters VGLUT1 and VGLUT2 in the rat dental pulp and trigeminal ganglion following inflammation. PLoS One 2014; 9:e109723. [PMID: 25290694 PMCID: PMC4188624 DOI: 10.1371/journal.pone.0109723] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 09/04/2014] [Indexed: 01/07/2023] Open
Abstract
Background There is increasing evidence that peripheral glutamate signaling mechanism is involved in the nociceptive transmission during pathological conditions. However, little is known about the glutamate signaling mechanism and related specific type of vesicular glutamate transporter (VGLUT) in the dental pulp following inflammation. To address this issue, we investigated expression and protein levels of VGLUT1 and VGLUT2 in the dental pulp and trigeminal ganglion (TG) following complete Freund’s adjuvant (CFA) application to the rat dental pulp by light microscopic immunohistochemistry and Western blot analysis. Results The density of VGLUT2− immunopositive (+) axons in the dental pulp and the number of VGLUT2+ soma in the TG increased significantly in the CFA-treated group, compared to control group. The protein levels of VGLUT2 in the dental pulp and TG were also significantly higher in the CFA-treated group than control group by Western blot analysis. The density of VGLUT1+ axons in the dental pulp and soma in the TG remained unchanged in the CFA-treated group. Conclusions These findings suggest that glutamate signaling that is mediated by VGLUT2 in the pulpal axons may be enhanced in the inflamed dental pulp, which may contribute to pulpal axon sensitization leading to hyperalgesia following inflammation.
Collapse
|
37
|
Harris KD, Weiss M, Zahavi A. Why are neurotransmitters neurotoxic? An evolutionary perspective. F1000Res 2014; 3:179. [PMID: 25580225 PMCID: PMC4288432 DOI: 10.12688/f1000research.4828.2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2014] [Indexed: 02/02/2023] Open
Abstract
In the CNS, minor changes in the concentration of neurotransmitters such as glutamate or dopamine can lead to neurodegenerative diseases. We present an evolutionary perspective on the function of neurotransmitter toxicity in the CNS. We hypothesize that neurotransmitters are selected because of their toxicity, which serves as a test of neuron quality and facilitates the selection of neuronal pathways. This perspective may offer additional explanations for the reduction of neurotransmitter concentration in the CNS with age, and suggest an additional role for the blood-brain barrier. It may also suggest a connection between the specific toxicity of the neurotransmitters released in a specific region of the CNS, and elucidate their role as chemicals that are optimal for testing the quality of cells in that region.
Collapse
Affiliation(s)
- Keith D Harris
- Department of Zoology, Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Meital Weiss
- Department of Zoology, Tel-Aviv University, Tel Aviv, 69978, Israel
| | - Amotz Zahavi
- Department of Zoology, Tel-Aviv University, Tel Aviv, 69978, Israel ; Sagol School of Neuroscience, Tel-Aviv University, Tel Aviv, 69978, Israel
| |
Collapse
|
38
|
Morales M, Root DH. Glutamate neurons within the midbrain dopamine regions. Neuroscience 2014; 282:60-8. [PMID: 24875175 DOI: 10.1016/j.neuroscience.2014.05.032] [Citation(s) in RCA: 182] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/15/2014] [Accepted: 05/18/2014] [Indexed: 12/26/2022]
Abstract
Midbrain dopamine systems play important roles in Parkinson's disease, schizophrenia, addiction, and depression. The participation of midbrain dopamine systems in diverse clinical contexts suggests these systems are highly complex. Midbrain dopamine regions contain at least three neuronal phenotypes: dopaminergic, GABAergic, and glutamatergic. Here, we review the locations, subtypes, and functions of glutamatergic neurons within midbrain dopamine regions. Vesicular glutamate transporter 2 (VGluT2) mRNA-expressing neurons are observed within each midbrain dopamine system. Within rat retrorubral field (RRF), large populations of VGluT2 neurons are observed throughout its anteroposterior extent. Within rat substantia nigra pars compacta (SNC), VGluT2 neurons are observed centrally and caudally, and are most dense within the laterodorsal subdivision. RRF and SNC rat VGluT2 neurons lack tyrosine hydroxylase (TH), making them an entirely distinct population of neurons from dopaminergic neurons. The rat ventral tegmental area (VTA) contains the most heterogeneous populations of VGluT2 neurons. VGluT2 neurons are found in each VTA subnucleus but are most dense within the anterior midline subnuclei. Some subpopulations of rat VGluT2 neurons co-express TH or glutamic acid decarboxylase (GAD), but most of the VGluT2 neurons lack TH or GAD. Different subsets of rat VGluT2-TH neurons exist based on the presence or absence of vesicular monoamine transporter 2, dopamine transporter, or D2 dopamine receptor. Thus, the capacity by which VGluT2-TH neurons may release dopamine will differ based on their capacity to accumulate vesicular dopamine, uptake extracellular dopamine, or be autoregulated by dopamine. Rat VTA VGluT2 neurons exhibit intrinsic VTA projections and extrinsic projections to the accumbens and to the prefrontal cortex. Mouse VTA VGluT2 neurons project to accumbens shell, prefrontal cortex, ventral pallidum, amygdala, and lateral habenula. Given their molecular diversity and participation in circuits involved in addiction, we hypothesize that individual VGluT2 subpopulations of neurons play unique roles in addiction and other disorders.
Collapse
Affiliation(s)
- M Morales
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard, Suite 200, Baltimore, MD 21224, United States.
| | - D H Root
- Neuronal Networks Section, Integrative Neuroscience Research Branch, National Institute on Drug Abuse, 251 Bayview Boulevard, Suite 200, Baltimore, MD 21224, United States
| |
Collapse
|
39
|
Omote H, Moriyama Y. Vesicular neurotransmitter transporters: an approach for studying transporters with purified proteins. Physiology (Bethesda) 2014; 28:39-50. [PMID: 23280356 DOI: 10.1152/physiol.00033.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vesicular storage and subsequent release of neurotransmitters are the key processes of chemical signal transmission. In this process, vesicular neurotransmitter transporters are responsible for loading the signaling molecules. The use of a "clean biochemical" approach with purified, recombinant transporters has helped in the identification of novel vesicular neurotransmitter transporters and in the analysis of the control of signal transmission.
Collapse
Affiliation(s)
- Hiroshi Omote
- Department of Membrane Biochemistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
40
|
Jenstad M, Chaudhry FA. The Amino Acid Transporters of the Glutamate/GABA-Glutamine Cycle and Their Impact on Insulin and Glucagon Secretion. Front Endocrinol (Lausanne) 2013; 4:199. [PMID: 24427154 PMCID: PMC3876026 DOI: 10.3389/fendo.2013.00199] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/16/2013] [Indexed: 12/21/2022] Open
Abstract
Intercellular communication is pivotal in optimizing and synchronizing cellular responses to keep homeostasis and to respond adequately to external stimuli. In the central nervous system (CNS), glutamatergic and GABAergic signals are postulated to be dependent on the glutamate/GABA-glutamine cycle for vesicular loading of neurotransmitters, for inactivating the signal and for the replenishment of the neurotransmitters. Islets of Langerhans release the hormones insulin and glucagon, but share similarities with CNS cells in for example transcriptional control of development and differentiation, and chromatin methylation. Interestingly, CNS proteins involved in secretion of the neurotransmitters and emitting their responses as well as the regulation of these processes, are also found in islet cells. Moreover, high levels of glutamate, GABA, and glutamine and their respective vesicular and plasma membrane transporters have been shown in the islet cells and there is emerging support for these amino acids and their transporters playing important roles in the maturation and secretion of insulin and glucagon. In this review, we will discuss the feasibility of recent data in the field in relation to the biophysical properties of the transporters (Slc1, Slc17, Slc32, and Slc38) and physiology of hormone secretion in islets of Langerhans.
Collapse
Affiliation(s)
- Monica Jenstad
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
- Centre for Cancer Biomedicine, University of Oslo, Oslo, Norway
- *Correspondence: Monica Jenstad, Institute for Medical Informatics, Oslo University Hospital, Radiumhospitalet, PO Box 4953 Nydalen, Oslo NO-0424, Norway e-mail:
| | - Farrukh Abbas Chaudhry
- Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- The Biotechnology Centre of Oslo, University of Oslo, Oslo, Norway
| |
Collapse
|
41
|
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
|
42
|
Guardado-Mendoza R, Jimenez-Ceja L, Majluf-Cruz A, Kamath S, Fiorentino TV, Casiraghi F, Velazquez AOC, DeFronzo RA, Dick E, Davalli A, Folli F. Impact of obesity severity and duration on pancreatic β- and α-cell dynamics in normoglycemic non-human primates. Int J Obes (Lond) 2013; 37:1071-8. [PMID: 23229736 PMCID: PMC3906680 DOI: 10.1038/ijo.2012.205] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 11/05/2012] [Accepted: 11/08/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Obesity is associated with high insulin and glucagon plasma levels. Enhanced β-cell function and β-cell expansion are responsible for insulin hypersecretion. It is unknown whether hyperglucagonemia is due to α-cell hypersecretion or to an increase in α-cell mass. In this study, we investigated the dynamics of the β-cell and α-cell function and mass in pancreas of obese normoglycemic baboons. METHODS Pancreatic β- and α-cell volumes were measured in 51 normoglycemic baboons divided into six groups according to overweight severity or duration. Islets morphometric parameters were correlated to overweight and to diverse metabolic and laboratory parameters. RESULTS Relative α-cell volume (RαV) and relative islet α-cell volume (RIαV) increased significantly with both overweight duration and severity. Conversely, in spite of the induction of insulin resistance, overweight produced only modest effects on relative β-cell volume (RβV) and relative islet β-cell volume (RIβV). Of note, RIβV did not increase neither with overweight duration nor with overweight severity, supposedly because of the concomitant, greater increase in RIαV. Baboons' body weights correlated with serum levels of interleukin-6 and tumor necrosis factor-α soluble receptors, demonstrating that overweight induces abnormal activation of the signaling of two cytokines known to impact differently β- and α-cell viability and replication. CONCLUSION In conclusion, overweight and insulin resistance induce in baboons a significant increase in α-cell volumes (RαV, RIαV), whereas have minimal effects on the β cells. This study suggests that an increase in the α-cell mass may precede the loss of β cells and the transition to overt hyperglycemia and diabetes.
Collapse
Affiliation(s)
- R Guardado-Mendoza
- Department of Medicine, Diabetes Division, University of Texas Health Science Center at San Antonio, TX, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Cuttitta CM, Guariglia SR, Idrissi AE, L’Amoreaux WJ. Taurine’s Effects on the Neuroendocrine Functions of Pancreatic β Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 775:299-310. [DOI: 10.1007/978-1-4614-6130-2_25] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
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]
|
45
|
Caicedo A. Paracrine and autocrine interactions in the human islet: more than meets the eye. Semin Cell Dev Biol 2012; 24:11-21. [PMID: 23022232 DOI: 10.1016/j.semcdb.2012.09.007] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 09/19/2012] [Indexed: 12/20/2022]
Abstract
The pancreatic islet secretes the hormones insulin and glucagon to regulate glucose metabolism. To generate an adequate secretory response, islet endocrine cells must receive multiple regulatory signals relaying information about changes in the internal and external environments. Islet cells also need to be made aware about the functional status of neighboring cells through paracrine interactions. All this information is used to orchestrate a hormonal response that contributes to glucose homeostasis. Several neurotransmitters have been proposed to work as paracrine signals in the islet. Most of these, however, have yet to meet the criteria to be considered bona fide paracrine signals, in particular in human islets. Here, we review recent findings describing autocrine and paracrine signaling mechanisms in human islets. These recent results are showing an increasingly complex picture of paracrine interactions in the human islet and emphasize that results from other species cannot be readily extrapolated to the human context. Investigators are unveiling new signaling mechanisms or finding new roles for known paracrine signals in human islets. While it is too early to provide a synthesis, the field of islet research is defining the paracrine and autocrine components that will be used to generate models about how islet function is regulated. Meanwhile, the identified signaling pathways can be proposed as therapeutic targets for treating diabetes, a devastating disease affecting millions worldwide.
Collapse
Affiliation(s)
- Alejandro Caicedo
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
46
|
Gupta R, Palchaudhuri S, Chattopadhyay D. Glutamate induces neutrophil cell migration by activating class I metabotropic glutamate receptors. Amino Acids 2012; 44:757-67. [DOI: 10.1007/s00726-012-1400-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 09/05/2012] [Indexed: 12/11/2022]
|
47
|
Shafik AN. Effects of topiramate on diabetes mellitus induced by streptozotocin in rats. Eur J Pharmacol 2012; 684:161-167. [PMID: 22498001 DOI: 10.1016/j.ejphar.2012.03.042] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 03/14/2012] [Accepted: 03/23/2012] [Indexed: 12/18/2022]
Abstract
Topiramate currently approved for marketing as antiepileptic drug also possesses anti-diabetic activity. The aim of this study was to determine the antidiabetic effect of topiramate in a rat model of diabetes mellitus. Diabetes was induced by a single injection of streptozotocin to fasted rats. Diabetic animals were divided into untreated; insulin treated; topiramate treated with 25, 50 and 100 mg/kg; and combined insulin plus topiramate treatment in the previous doses. All medications were given once daily started after the rise of blood glucose for three weeks. Control rats were divided into untreated; vehicle treated and rats given topiramate in the previous doses. Body weight, blood-glucose and insulin levels were measured. Histopathological examination, immunohistochemical and morphometric studies of islets of the pancreas were done. Topiramate 50 and 100mg/kg resulted in a significant decrease in the blood glucose and increase in the insulin levels as well as the number of islets and the count and mass of beta cells. Combined treatment to diabetic rats with insulin and topiramate induced a better response than either alone. Further experimental and clinical studies are needed to explore the different mechanisms of action of topiramate as antidiabetic both in insulin dependent and non-insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- Amani Nabil Shafik
- Department of Pharmacology, Faculty of Medicine, Cairo University, Egypt.
| |
Collapse
|
48
|
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
|
49
|
Wu ZY, Zhu LJ, Zou N, Bombek LK, Shao CY, Wang N, Wang XX, Liang L, Xia J, Rupnik M, Shen Y. AMPA receptors regulate exocytosis and insulin release in pancreatic β cells. Traffic 2012; 13:1124-39. [PMID: 22540213 DOI: 10.1111/j.1600-0854.2012.01373.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Revised: 04/25/2012] [Accepted: 04/27/2012] [Indexed: 01/06/2023]
Abstract
Ionotropic glutamate receptors (iGluRs) are expressed in islets and insulinoma cells and involved in insulin secretion. However, the exact roles that iGluRs play in β cells remain unclear. Here, we demonstrated that GluR2-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) were expressed in mouse β cells. Glutamate application increased both cytosolic calcium and the number of docked insulin-containing granules, which resulted in augmentation of depolarization-induced exocytosis and high-glucose-stimulated insulin release. While glutamate application directly depolarized β cells, it also induced an enormous depolarization when K(ATP) channels were available. Glutamate application reduced the conductance of K(ATP) channels and increased voltage oscillations. Moreover, actions of AMPARs were absent in Kir6.2 knock-out mice. The effects of AMPARs on K(ATP) channels were mediated by cytosolic cGMP. Taken together, our experiments uncovered a novel mechanism by which AMPARs participate in insulin release.
Collapse
Affiliation(s)
- Zhen-Yong Wu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, P. R. China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
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
|