1
|
Sharma A, Sah N, Sharma R, Vyas P, Liyanage W, Kannan S, Kannan RM. Development of a novel glucose-dendrimer based therapeutic targeting hyperexcitable neurons in neurological disorders. Bioeng Transl Med 2024; 9:e10655. [PMID: 39553433 PMCID: PMC11561801 DOI: 10.1002/btm2.10655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 11/19/2024] Open
Abstract
Neuronal hyperexcitability and excitotoxicity lies at the core of debilitating brain disorders such as epilepsy and traumatic brain injury, culminating in neuronal death and compromised brain function. Overcoming this challenge requires a unique approach that selectively restores normal neuronal activity and rescues neurons from impending damage. However, delivering drugs selectively to hyperexcitable neurons has been a challenge, even upon local administration. Here, we demonstrate the remarkable ability of a novel, scalable, generation-two glucose-dendrimer (GD2) made primarily of glucose and ethylene glycol building blocks, to specifically target hyperexcitable neurons in primary culture, ex vivo acute brain slices, and in vivo mouse models of acute seizures. Pharmacology experiments in ex vivo brain slices suggest GD2 uptake in neurons is mediated through glucose transporters (GLUT and SGLT). Inspired by these findings, we conjugated GD2 with a potent anti-epileptic drug, valproic acid (GD2-VPA), for efficacy studies in the pilocarpine-mouse model of seizure. When delivered intranasally, GD2-VPA significantly decreased the seizure-severity. In summary, our findings demonstrate the unique selectivity of glucose dendrimers in targeting hyperexcitable neurons, even upon intranasal delivery, laying the foundation for neuron-specific therapies for the precise protection and restoration of neuronal function, for targeted neuroprotection.
Collapse
Affiliation(s)
- Anjali Sharma
- Center for Nanomedicine at the Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Present address:
Department of ChemistryWashington State UniversityPullmanWashingtonUSA
| | - Nirnath Sah
- Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Rishi Sharma
- Center for Nanomedicine at the Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Present address:
Department of ChemistryWashington State UniversityPullmanWashingtonUSA
| | - Preeti Vyas
- Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Wathsala Liyanage
- Center for Nanomedicine at the Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Sujatha Kannan
- Anesthesiology and Critical Care MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Rangaramanujam M. Kannan
- Center for Nanomedicine at the Wilmer Eye InstituteJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
2
|
Bonamy C, Pesnel S, Ben Haddada M, Gorgette O, Schmitt C, Morel AL, Sauvonnet N. Impact of Green Gold Nanoparticle Coating on Internalization, Trafficking, and Efficiency for Photothermal Therapy of Skin Cancer. ACS OMEGA 2023; 8:4092-4105. [PMID: 36743010 PMCID: PMC9893490 DOI: 10.1021/acsomega.2c07054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/29/2022] [Indexed: 06/18/2023]
Abstract
Skin cancer is a global health issue and mainly composed of melanoma and nonmelanoma cancers. For the first clinical proof of concept on humans, we decided to study good prognosis skin cancers, i.e., carcinoma basal cell. In UE, the first-line treatment remains surgical resection, healing most of the tumors, but presents aesthetic disadvantages with a high reoccurrence rate on exposed areas. Moreover, the therapeutic indications could extend to melanoma and metastasis, which is a different medical strategy that could combine this treatment. Indeed, patients with late-stage melanoma are in a therapeutic impasse, despite recent targeted and immunological therapies. Photothermal therapy using gold nanoparticles is the subject of many investigations due to their strong potential to treat cancers by physical, thermal destruction. We developed gold nanoparticles synthesized by green chemistry (gGNPs), using endemic plant extract from Reunion Island, which have previously showed their efficiency at a preclinical stage. Here, we demonstrate that these gGNPs are less cytotoxic than gold nanoparticles synthesized by Turkevich's method. Furthermore, our work describes the optimization of gGNP coating and stabilization, also taking into consideration the gGNP path in cells (endocytosis, intracellular trafficking, and exocytosis), their specificity toward cancerous cells, their cytotoxicity, and their in vivo efficiency. Finally, based on the metabolic switch of cancerous cells overexpressing Glut transporters in skin cancers, we demonstrated that glucose-stabilized gGNP (gGNP@G) enables a quick internalization, fourfold higher in cancerous cells in contrast to healthy cells with no side cytotoxicity, which is particularly relevant to target and treat cancer.
Collapse
Affiliation(s)
- Clément Bonamy
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sabrina Pesnel
- Torskal, 2 rue Maxime Rivière, 97490 Sainte-Clotilde, France
| | | | - Olivier Gorgette
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | - Christine Schmitt
- Ultrastructural
BioImaging, Institut Pasteur, Université
Paris Cité, 75015 Paris, France
| | | | - Nathalie Sauvonnet
- Group
Intracellular Trafficking and Tissue Homeostasis, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
3
|
The aminopeptidase B (Ap-B) is phosphorylated in HEK293 cells. Biochimie 2022; 201:204-212. [PMID: 35952945 DOI: 10.1016/j.biochi.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/22/2022]
Abstract
Proteolysis is a post-translational modification (PTM) that affects the whole proteome. First regarded as only destructive, it is more precise than expected. It is finely regulated by other PTMs like phosphorylation. Aminopeptidase B (Ap-B), a M1 metallopeptidase, hydrolyses the peptide bond on the carbonyl side of basic residues at the NH2-terminus of peptides. 2D electrophoresis (2DE) was used to show that Ap-B is modified by phosphorylation. Detection of Ap-B by western blot after 2DE reveals several isoforms with different isoelectric points. Using alkaline phosphatase, Pro-Q Diamond phosphorylation-specific dye and kinase-specific inhibitors, we confirmed that Ap-B is phosphorylated. Phosphorylation can alter the structure of proteins leading to changes in their activity, localization, stability and association with other interacting molecules. We showed that Ap-B phosphorylation might delay its turnover. Our study illustrates the central role of the crosstalk between kinases and proteases in the regulation of many biological processes.
Collapse
|
4
|
Zhu J, Zhang Z, Jia J, Wang L, Yang Q, Wang Y, Chen C. Sevoflurane Induces Learning and Memory Impairment in Young Mice Through a Reduction in Neuronal Glucose Transporter 3. Cell Mol Neurobiol 2020; 40:879-895. [PMID: 31884568 PMCID: PMC7295720 DOI: 10.1007/s10571-019-00779-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022]
Abstract
Sevoflurane, which is widely used in paediatric anaesthesia, induces neural apoptosis in the developing brain and cognitive impairment in young mammals. Glucose hypometabolism is the key pathophysiological modulator of cognitive dysfunction. However, the effects and mechanism of sevoflurane on cerebral glucose metabolism after its use as an anaesthetic and its complete elimination are still unknown. We therefore investigated the influence of sevoflurane on neuronal glucose transporter isoform 3 (GLUT3) expression, glucose metabolism and apoptosis in vivo and in vitro and on neurocognitive function in young mice 24 h after the third exposure to sevoflurane. Postnatal day 14 (P14) mice and neural cells were exposed to 3% sevoflurane 2 h daily for three days. We found that sevoflurane anaesthesia decreased GLUT3 gene and protein expression in the hippocampus and temporal lobe, consistent with a decrease in glucose metabolism in the hippocampus and temporal lobe observed by [18F] fluorodeoxyglucose positron emission tomography (18F-FDG PET). Moreover, sevoflurane anaesthesia increased the number of TUNEL-positive cells and the levels of Bax, cleaved caspase 3 and cleaved PARP and reduced Bcl-2 levels in the hippocampus and temporal lobe. Young mice exposed to sevoflurane multiple times also showed learning and memory impairment. In addition, sevoflurane inhibited GLUT3 expression in primary hippocampal neurons and PC12 cells. GLUT3 overexpression in cultured neurons ameliorated the sevoflurane-induced decrease in glucose utilization and increase in the apoptosis rate. These data indicate that GLUT3 deficiency may contribute to sevoflurane-induced learning and memory deficits in young mice.
Collapse
Affiliation(s)
- Jinpiao Zhu
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Zongze Zhang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Junke Jia
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Lirong Wang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Qiuyue Yang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Yanlin Wang
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China
| | - Chang Chen
- Department of Anaesthesiology, Zhongnan Hospital, Wuhan University, East Lake Road, Wuhan, 430071, Hubei, China.
| |
Collapse
|
5
|
Ziegler GC, Almos P, McNeill RV, Jansch C, Lesch KP. Cellular effects and clinical implications of SLC2A3 copy number variation. J Cell Physiol 2020; 235:9021-9036. [PMID: 32372501 DOI: 10.1002/jcp.29753] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/04/2020] [Accepted: 04/21/2020] [Indexed: 12/19/2022]
Abstract
SLC2A3 encodes the predominantly neuronal glucose transporter 3 (GLUT3), which facilitates diffusion of glucose across plasma membranes. The human brain depends on a steady glucose supply for ATP generation, which consequently fuels critical biochemical processes, such as axonal transport and neurotransmitter release. Besides its role in the central nervous system, GLUT3 is also expressed in nonneural organs, such as the heart and white blood cells, where it is equally involved in energy metabolism. In cancer cells, GLUT3 overexpression contributes to the Warburg effect by answering the cell's increased glycolytic demands. The SLC2A3 gene locus at chromosome 12p13.31 is unstable and prone to non-allelic homologous recombination events, generating multiple copy number variants (CNVs) of SLC2A3 which account for alterations in SLC2A3 expression. Recent associations of SLC2A3 CNVs with different clinical phenotypes warrant investigation of the potential influence of these structural variants on pathomechanisms of neuropsychiatric, cardiovascular, and immune diseases. In this review, we accumulate and discuss the evidence how SLC2A3 gene dosage may exert diverse protective or detrimental effects depending on the pathological condition. Cellular states which lead to increased energetic demand, such as organ development, proliferation, and cellular degeneration, appear particularly susceptible to alterations in SLC2A3 copy number. We conclude that better understanding of the impact of SLC2A3 variation on disease etiology may potentially provide novel therapeutic approaches specifically targeting this GLUT.
Collapse
Affiliation(s)
- Georg C Ziegler
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany.,Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Würzburg, Germany
| | - Peter Almos
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany.,Department of Psychiatry, University of Szeged, Hungary
| | - Rhiannon V McNeill
- Department of Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University of Würzburg, Germany
| | - Charline Jansch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany
| | - Klaus-Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Germany.,Department of Psychiatry and Neuropsychology, School of Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
6
|
Dehydroascorbic Acid Promotes Cell Death in Neurons Under Oxidative Stress: a Protective Role for Astrocytes. Mol Neurobiol 2015; 53:5847-5863. [PMID: 26497038 DOI: 10.1007/s12035-015-9497-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022]
Abstract
Ascorbic acid (AA), the reduced form of vitamin C, is incorporated into neurons via the sodium ascorbate co-transporter SVCT2. However, this transporter is not expressed in astrocytes, which take up the oxidized form of vitamin C, dehydroascorbic acid (DHA), via the facilitative hexose transporter GLUT1. Therefore, neuron and astrocyte interactions are thought to mediate vitamin C recycling in the nervous system. Although astrocytes are essential for the antioxidant defense of neurons under oxidative stress, a condition in which a large amount of ROS is generated that may favor the extracellular oxidation of AA and the subsequent neuronal uptake of DHA via GLUT3, potentially increasing oxidative stress in neurons. This study analyzed the effects of oxidative stress and DHA uptake on neuronal cell death in vitro. Different analyses revealed the presence of the DHA transporters GLUT1 and GLUT3 in Neuro2a and HN33.11 cells and in cortical neurons. Kinetic analyses confirmed that all cells analyzed in this study possess functional GLUTs that take up 2-deoxyglucose and DHA. Thus, DHA promotes the death of stressed neuronal cells, which is reversed by incubating the cells with cytochalasin B, an inhibitor of DHA uptake by GLUT1 and GLUT3. Additionally, the presence of glial cells (U87 and astrocytes), which promote DHA recycling, reverses the observed cell death of stressed neurons. Taken together, these results indicate that DHA promotes the death of stressed neurons and that astrocytes are essential for the antioxidative defense of neurons. Thus, the astrocyte-neuron interaction may function as an essential mechanism for vitamin C recycling, participating in the antioxidative defense of the brain.
Collapse
|
7
|
Chen Y, Shin BC, Thamotharan S, Devaskar SU. Creb1-Mecp2-(m)CpG complex transactivates postnatal murine neuronal glucose transporter isoform 3 expression. Endocrinology 2013; 154:1598-611. [PMID: 23493374 PMCID: PMC3602632 DOI: 10.1210/en.2012-2076] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The murine neuronal facilitative glucose transporter isoform 3 (Glut3) is developmentally regulated, peaking in expression at postnatal day (PN)14. In the present study, we characterized a canonical CpG island spanning the 5'-flanking region of the glut3 gene. Methylation-specific PCR and bisulfite sequencing identified methylation of this CpG ((m)CpG) island of the glut3 gene, frequency of methylation increasing 2.5-fold with a 1.6-fold increase in DNA methyl transferase 3a concentrations noted with advancing postnatal age (PN14 vs PN3). 5'-flanking region of glut3-luciferase reporter transient transfection in HT22 hippocampal neurons demonstrated that (m)CpGs inhibit glut3 transcription. Contrary to this biological function, glut3 expression rises synchronously with (m)CpGs in PN14 vs PN3 neurons. Chromatin immunoprecipitation (IP) revealed that methyl-CpG binding protein 2 (Mecp2) bound the glut3-(m)CpGs. Depending on association with specific coregulators, Mecp2, a dual regulator of gene transcription, may repress or activate a downstream gene. Sequential chromatin IP uncovered the glut3-(m)CpGs to bind Mecp2 exponentially upon recruitment of Creb1 rather than histone deacetylase 1. Co-IP and coimmunolocalization confirmed that Creb1 associated with Mecp2 and cotransfection with glut3-(m)CpG in HT22 cells enhanced glut3 transcription. Separate 5-aza-2'-deoxycytidine pretreatment or in combination with trichostatin A reduced (m)CpG and specific small interference RNAs targeting Mecp2 and Creb1 separately or together depleting Mecp2 and/or Creb1 binding of glut3-(m)CpGs reduced glut3 expression in HT22 cells. We conclude that Glut3 is a methylation-sensitive neuronal gene that recruits Mecp2. Recruitment of Creb1-Mecp2 by glut3-(m)CpG contributes towards transactivation, formulating an escape from (m)CpG-induced gene suppression, and thereby promoting developmental neuronal glut3 gene transcription and expression.
Collapse
Affiliation(s)
- Yongjun Chen
- Department of Pediatrics, Division of Neonatology and Developmental Biology, Neonatal Research Center, David Geffen School of Medicine University of California LosAngeles, Los Angeles, California 90095-1752, USA
| | | | | | | |
Collapse
|
8
|
Ronquist KG, Ek B, Stavreus-Evers A, Larsson A, Ronquist G. Human prostasomes express glycolytic enzymes with capacity for ATP production. Am J Physiol Endocrinol Metab 2013; 304:E576-82. [PMID: 23341497 DOI: 10.1152/ajpendo.00511.2012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Prostasomes are prostate-derived, exosome-like microvesicles that transmit signaling complexes between the acinar epithelial cells of the prostate and sperm cells. The vast majority of prostasomes have a diameter of 30-200 nm, and they are generally surrounded by a classical membrane bilayer. Using a selected proteomic approach, it became increasingly clear that prostasomes harbor distinct subsets of proteins that may be linked to adenosine triphosphate (ATP) metabolic turnover that in turn might be of importance in the role of prostasomes as auxiliary instruments in the fertilization process. Among the 21 proteins identified, most of the enzymes of anaerobic glycolysis were represented, and three of the glycolytic enzymes present are among the top 10 proteins found in most exosomes, once again linking prostasomes to the exosome family. Other prostasomal enzymes involved in ATP turnover were adenylate kinase, ATPase, 5'-nucleotidase, and hexose transporters. The identified enzymes in their prostasomal context were operational for ATP formation when supplied with substrates. The net ATP production was low due to a high prostasomal ATPase activity that could be partially inhibited by vanadate that was utilized to profile the ATP-forming ability of prostasomes. Glucose and fructose were equivalent as glycolytic substrates for prostasomal ATP formation, and the enzymes involved were apparently surface located on prostasomes, since an alternative substrate not being membrane permeable (glyceraldehyde 3-phosphate) was operative, too. There is no clear-cut function linked to this subset of prostasomal proteins, but some possible roles are discussed.
Collapse
Affiliation(s)
- K Göran Ronquist
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
9
|
Pham VL, Gouzy-Darmon C, Pernier J, Hanquez C, Hook V, Beinfeld MC, Nicolas P, Etchebest C, Foulon T, Cadel S. Mutation in the substrate-binding site of aminopeptidase B confers new enzymatic properties. Biochimie 2011; 93:730-41. [DOI: 10.1016/j.biochi.2010.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 12/24/2010] [Indexed: 02/03/2023]
|
10
|
Núñez E, Pérez-Siles G, Rodenstein L, Alonso-Torres P, Zafra F, Jiménez E, Aragón C, López-Corcuera B. Subcellular localization of the neuronal glycine transporter GLYT2 in brainstem. Traffic 2009; 10:829-43. [PMID: 19374720 DOI: 10.1111/j.1600-0854.2009.00911.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The neuronal glycine transporter GLYT2 belongs to the neurotransmitter:sodium:symporter (NSS) family and removes glycine from the synaptic cleft, thereby aiding the termination of the glycinergic signal and achieving the reloading of the presynaptic terminal. The task fulfilled by this transporter is fine tuned by regulating both transport activity and intracellular trafficking. Different stimuli such as neuronal activity or protein kinase C (PKC) activation can control GLYT2 surface levels although the intracellular compartments where GLYT2 resides are largely unknown. Here, by biochemical and immunological techniques in combination with electron and confocal microscopy, we have investigated the subcellular distribution of GLYT2 in rat brainstem tissue, and characterized the vesicles that contain the transporter. GLYT2 is shown to be present in small and larger vesicles that contain the synaptic vesicle protein synaptophysin, the recycling endosome small GTPase Rab11, and in the larger vesicle population, the vesicular inhibitory amino acid transporter VIAAT. Rab5A, the GABA transporter GAT1, synaptotagmin2 and synaptobrevin2 (VAMP2) were not present. Coexpression of a Rab11 dominant negative mutant with recombinant GLYT2 impaired transporter trafficking and glycine transport. Dual immunogold labeling of brainstem synaptosomes showed a very close proximity of GLYT2 and Rab11. Therefore, the intracellular GLYT2 resides in a subset of endosomal membranes and may traffic around several compartments, mainly Rab11-positive endosomes.
Collapse
Affiliation(s)
- Enrique Núñez
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC). Universidad Autónoma de Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Pham VL, Cadel MS, Gouzy-Darmon C, Hanquez C, Beinfeld MC, Nicolas P, Etchebest C, Foulon T. Aminopeptidase B, a glucagon-processing enzyme: site directed mutagenesis of the Zn2+-binding motif and molecular modelling. BMC BIOCHEMISTRY 2007; 8:21. [PMID: 17974014 PMCID: PMC2241622 DOI: 10.1186/1471-2091-8-21] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Accepted: 10/31/2007] [Indexed: 12/18/2022]
Abstract
BACKGROUND Aminopeptidase B (Ap-B; EC 3.4.11.6) catalyzes the cleavage of basic residues at the N-terminus of peptides and processes glucagon into miniglucagon. The enzyme exhibits, in vitro, a residual ability to hydrolyze leukotriene A4 into the pro-inflammatory lipid mediator leukotriene B4. The potential bi-functional nature of Ap-B is supported by close structural relationships with LTA4 hydrolase (LTA4H ; EC 3.3.2.6). A structure-function analysis is necessary for the detailed understanding of the enzymatic mechanisms of Ap-B and to design inhibitors, which could be used to determine the complete in vivo functions of the enzyme. RESULTS The rat Ap-B cDNA was expressed in E. coli and the purified recombinant enzyme was characterized. 18 mutants of the H325EXXHX18E348 Zn2+-binding motif were constructed and expressed. All mutations were found to abolish the aminopeptidase activity. A multiple alignment of 500 sequences of the M1 family of aminopeptidases was performed to identify 3 sub-families of exopeptidases and to build a structural model of Ap-B using the x-ray structure of LTA4H as a template. Although the 3D structures of the two enzymes resemble each other, they differ in certain details. The role that a loop, delimiting the active center of Ap-B, plays in discriminating basic substrates, as well as the function of consensus motifs, such as RNP1 and Armadillo domain are discussed. Examination of electrostatic potentials and hydrophobic patches revealed important differences between Ap-B and LTA4H and suggests that Ap-B is involved in protein-protein interactions. CONCLUSION Alignment of the primary structures of the M1 family members clearly demonstrates the existence of different sub-families and highlights crucial residues in the enzymatic activity of the whole family. E. coli recombinant enzyme and Ap-B structural model constitute powerful tools for investigating the importance and possible roles of these conserved residues in Ap-B, LTA4H and M1 aminopeptidase catalytic sites and to gain new insight into their physiological functions. Analysis of Ap-B structural model indicates that several interactions between Ap-B and proteins can occur and suggests that endopeptidases might form a complex with Ap-B during hormone processing.
Collapse
Affiliation(s)
- Viet-Laï Pham
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| | - Marie-Sandrine Cadel
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| | - Cécile Gouzy-Darmon
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| | - Chantal Hanquez
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| | - Margery C Beinfeld
- Department of Pharmacology and Experimental Therapeutics, Tufts University, School of Medicine, Boston, MA 02111, USA
| | - Pierre Nicolas
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| | - Catherine Etchebest
- Université Denis Diderot-Paris7, UMR S 726, INSERM, Laboratoire de Bioinformatique Génomique et Moléculaire, Paris, F-75251 France
| | - Thierry Foulon
- Université Pierre et Marie Curie-Paris6, FRE 2852 (CNRS), Protéines : Biochimie Structurale et Fonctionnelle, Structures et Fonctions des Aminopeptidases, Paris, F-75005 France
| |
Collapse
|
12
|
Sakyo T, Naraba H, Teraoka H, Kitagawa T. The intrinsic structure of glucose transporter isoforms Glut1 and Glut3 regulates their differential distribution to detergent-resistant membrane domains in nonpolarized mammalian cells. FEBS J 2007; 274:2843-53. [PMID: 17459098 DOI: 10.1111/j.1742-4658.2007.05814.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The hexose transporter family, which mediates facilitated uptake in mammalian cells, consists of more than 10 members containing 12 membrane-spanning segments with a single N-glycosylation site. We previously demonstrated that glucose transporter 1 is organized into a raft-like detergent-resistant membrane domain but that glucose transporter 3 distributes to fluid membrane domains in nonpolarized mammalian cells. In this study, we further examined the structural basis responsible for the distribution by using a series of chimeric constructs. Glucose transporter 1 and glucose transporter 3 with a FLAG-tagged N-terminus were expressed in detergent-resistant membranes and non-detergent-resistant membranes of CHO-K1 cells, respectively. Replacement of either the C-terminal or N-terminal cytosolic portion of FLAG-tagged glucose transporter 1 and glucose transporter 3 did not affect the membrane distribution. However, a critical sorting signal may exist within the N-terminal half of the isoforms without affecting transport activity and its inhibition by cytochalasin B. Further shortening of these regions altered the critical distribution, suggesting that a large proportion or several parts of the intrinsic structure, including the N-terminus of each isoform, are involved in the regulation.
Collapse
Affiliation(s)
- Tomoko Sakyo
- Pharmaceutical Research Center, School of Pharmacy, Iwate Medical University, Morioka, Iwate 028-3694, Japan
| | | | | | | |
Collapse
|
13
|
Barros LF, Bittner CX, Loaiza A, Porras OH. A quantitative overview of glucose dynamics in the gliovascular unit. Glia 2007; 55:1222-1237. [PMID: 17659523 DOI: 10.1002/glia.20375] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
While glucose is constantly being "pulled" into the brain by hexokinase, its flux across the blood brain barrier (BBB) is allowed by facilitative carriers of the GLUT family. Starting from the microscopic properties of GLUT carriers, and within the constraints imposed by the available experimental data, chiefly NMR spectroscopy, we have generated a numerical model that reveals several hidden features of glucose transport and metabolism in the brain. The half-saturation constant of glucose uptake into the brain (K(t)) is close to 8 mM. GLUT carriers at the BBB are symmetric, show accelerated-exchange, and a K(m) of zero-trans flux (K(zt)) close to 5 mM, determining a ratio of 3.6 between maximum transport rate and net glucose flux (T(max)/CMR(glc)). In spite of the low transporter occupancy, the model shows that for a stimulated hexokinase to pull more glucose into the brain, the number or activity of GLUT carriers must also increase, particularly at the BBB. The endothelium is therefore predicted to be a key modulated element for the fast control of energy metabolism. In addition, the simulations help to explain why mild hypoglycemia may be asymptomatic and reveal that [glucose](brain) (as measured by NMR) should be much more sensitive than glucose flux (as measured by PET) as an indicator of GLUT1 deficiency. In summary, available data from various sources has been integrated in a predictive model based on the microscopic properties of GLUT carriers.
Collapse
Affiliation(s)
- L F Barros
- Centro de Estudios Científicos (CECS), Av. Arturo Prat 514, Casilla 1469, Valdivia, Chile
| | - Carla X Bittner
- Centro de Estudios Científicos (CECS), Av. Arturo Prat 514, Casilla 1469, Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Anitsi Loaiza
- Centro de Estudios Científicos (CECS), Av. Arturo Prat 514, Casilla 1469, Valdivia, Chile
- Universidad Austral de Chile, Valdivia, Chile
| | - Omar H Porras
- Centro de Estudios Científicos (CECS), Av. Arturo Prat 514, Casilla 1469, Valdivia, Chile
| |
Collapse
|
14
|
Rauch MC, Ocampo ME, Bohle J, Amthauer R, Yáñez AJ, Rodríguez-Gil JE, Slebe JC, Reyes JG, Concha II. Hexose transporters GLUT1 and GLUT3 are colocalized with hexokinase I in caveolae microdomains of rat spermatogenic cells. J Cell Physiol 2006; 207:397-406. [PMID: 16419038 DOI: 10.1002/jcp.20582] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Postmeiotic spermatogenic cells, but not meiotic spermatogenic cells respond differentially with glucose-induced changes in [Ca2+]i indicating a differential transport of glucose via facilitative hexose transporters (GLUTs) specifically distributed in the plasma membrane. Several studies have indicated that plasma membrane in mammalian cells is not homogeneously organized, but contains specific microdomains known as detergent-resistant membrane domains (DRMDs), lipid rafts or caveolae. The association of these domains and GLUTs isoforms has not been characterized in spermatogenic cells. We analyzed the expression and function of GLUT1 and GLUT3 in isolated spermatocytes and spermatids. The results showed that spermatogenic cells express both glucose transporters, with spermatids exhibiting a higher affinity glucose transport system. In addition, spermatogenic cells express caveolin-1, and glucose transporters colocalize with caveolin-1 in caveolin-enriched membrane fractions. Experiments in which the integrity of caveolae was disrupted by pretreatment with methyl-beta-cyclodextrin, indicated that the involvement of cholesterol-enriched plasma membrane microdomains were involved in the localization of GLUTs and uptake of 2-deoxyglucose. We also observed cofractionation of GLUT3 and caveolin-1 in low-buoyant density membranes together with their shift to higher densities after methyl-beta-cyclodextrin treatment. GLUT1 was found in all fractions isolated. Immunofluorescent studies indicated that caveolin-1, GLUT1, and hexokinase I colocalize in spermatocytes while caveolin-1, GLUT3, and hexokinase I colocalize in spermatids. These findings suggest the presence of hexose transporters in DRMDs, and further support a role for intact caveolae or cholesterol-enriched membrane microdomains in relation to glucose uptake and glucose phosphorylation. The results would also explain the different glucose-induced changes in [Ca2+]i in both cells.
Collapse
|
15
|
Piesse C, Cadel S, Gouzy-Darmon C, Jeanny JC, Carrière V, Goidin D, Jonet L, Gourdji D, Cohen P, Foulon T. Expression of aminopeptidase B in the developing and adult rat retina. Exp Eye Res 2004; 79:639-48. [PMID: 15500823 DOI: 10.1016/j.exer.2004.06.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Accepted: 06/03/2004] [Indexed: 11/29/2022]
Abstract
Aminopeptidase B (Ap-B), a ubiquitous enzyme, catalyses the amino-terminal cleavage of basic residues of peptide or protein substrates, indicating a role in precursor processing. The physiological function of Ap-B still remains an open question, even though its activity suggests that it could be involved in inflammatory processes and proliferation of tumor cells. This study was conducted to determine the expression of Ap-B in the developing and adult retina as a path to envisage physiological roles of Ap-B. RT-PCR and in situ hybridization were used to detect expression of Ap-B mRNA and activity tests, Western blotting and immunofluorescence microscopy were performed to identify and localize the enzyme in the rat retina. These biochemical and morphological methods show that Ap-B is expressed in the retina from embryo to adult. Expression level is restricted to specific layers (pigmented epithelium, outer and inner plexiform layers and ganglion cell layer) and is developmentally regulated. Moreover, a preliminary analysis indicates that Ap-B, the glucose transporter GLUT3 and choline acetyltransferase (ChAT) share a similar expression pattern in retina. Altogether, Ap-B appears predominantly expressed in neuronal cells lying in retinal layers containing neuritic extensions and synaptic junctions. Such expression is up-regulated during ontogenesis allowing to hypothesized that Ap-B participates in processes accompanying retinal neuronal cell differentiation.
Collapse
Affiliation(s)
- Christophe Piesse
- Laboratoire de Biochimie des Signaux Régulateurs Cellulaires et Moléculaires, Unité Mixte de Recherche 7631, Université Pierre et Marie Curie-Centre National de la Recherche Scientifique, 96 Boulevard Raspail, 75006 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Cadel S, Gouzy-Darmon C, Petres S, Piesse C, Pham VL, Beinfeld MC, Cohen P, Foulon T. Expression and purification of rat recombinant aminopeptidase B secreted from baculovirus-infected insect cells. Protein Expr Purif 2004; 36:19-30. [PMID: 15177280 DOI: 10.1016/j.pep.2004.03.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Revised: 03/16/2004] [Indexed: 11/17/2022]
Abstract
Aminopeptidase B (Ap-B) is a ubiquitous enzyme and its physiological function still remains an open question. This Zn2+ -exopeptidase catalyzes the amino-terminal cleavage of basic residues of peptide or protein substrates, indicating a role in precursor processing. In addition, the enzyme exhibits a residual capacity to hydrolyze leukotriene A4 (LTA4) into the pro-inflammatory lipid mediator leukotriene B4 (LTB4) in vitro. This potential bi-functional nature of Ap-B is supported by a close structural relationship with LTA4 hydrolase, which hydrolyzes LTA4 into LTB4, in vivo, and exhibits an aminopeptidase activity, in vitro. Structural studies are necessary for the detailed understanding of the bi-functional enzymatic mechanism of Ap-B. In this study, we report cDNA cloning, baculovirus expression, and purification of the rat Ap-B (rAp-B). The Ap-B cDNA was constructed from extracted rat testes total RNA and introduced into the pBAC1 baculovirus transfer vector to generate recombinant baculoviruses. rAp-B expression, with or without COOH-hexahistidine tag, was tested in two different insect cell hosts (Sf9 and H5). The enzyme is secreted into the insect cell culture medium, which allowed a rapid purification of the protein. The His-tagged rAp-B was purified using metal affinity resin while the native recombinant rAp-B was partially purified using a single step DEAE Trisacryl ion exchange column. Although the recombinant rAp-B exhibits biochemical properties equivalent to those of the rat testes purified protein, the presence of the histidine-tag seems to partially inhibit the exopeptidase activity. However, this report shows that baculovirus-infected cells are a useful system to produce rat Ap-B for use in studying enzymatic mechanisms in vitro and 3D structure.
Collapse
Affiliation(s)
- Sandrine Cadel
- Laboratoire de Biochimie des Signaux Régulateurs Cellulaires et Moléculaires, FRE 2621, Université Pierre et Marie Curie--Centre National de la Recherche Scientifique, 96 Boulevard Raspail, 75006 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Uldry M, Thorens B. The SLC2 family of facilitated hexose and polyol transporters. Pflugers Arch 2004; 447:480-9. [PMID: 12750891 DOI: 10.1007/s00424-003-1085-0] [Citation(s) in RCA: 348] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2003] [Accepted: 04/04/2003] [Indexed: 12/18/2022]
Abstract
The SLC2 family of glucose and polyol transporters comprises 13 members, the glucose transporters (GLUT) 1-12 and the H(+)- myo-inositol cotransporter (HMIT). These proteins all contain 12 transmembrane domains with both the amino and carboxy-terminal ends located on the cytoplasmic side of the plasma membrane and a N-linked oligosaccharide side-chain located either on the first or fifth extracellular loop. Based on sequence comparison, the GLUT isoforms can be grouped into three classes: class I comprises GLUT1-4; class II, GLUT6, 8, 10, and 12 and class III, GLUT5, 7, 9, 11 and HMIT. Despite their sequence similarity and the presence of class-specific signature sequences, these transporters carry various hexoses and HMIT is a H(+)/ myo-inositol co-transporter. Furthermore, the substrate transported by some isoforms has not yet been identified. Tissue- and cell-specific expression of the well-characterized GLUT isoforms underlies their specific role in the control of whole-body glucose homeostasis. Numerous studies with transgenic or knockout mice indeed support an important role for these transporters in the control of glucose utilization, glucose storage and glucose sensing. Much remains to be learned about the transport functions of the recently discovered isoforms (GLUT6-13 and HMIT) and their physiological role in the metabolism of glucose, myo-inositol and perhaps other substrates.
Collapse
Affiliation(s)
- Marc Uldry
- Institute of Pharmacology and Toxicology, University of Lausanne, 27, Rue du Bugnon, 1005, Lausanne, Switzerland
| | | |
Collapse
|
18
|
Inukai K, Shewan AM, Pascoe WS, Katayama S, James DE, Oka Y. Carboxy terminus of glucose transporter 3 contains an apical membrane targeting domain. Mol Endocrinol 2003; 18:339-49. [PMID: 14605095 DOI: 10.1210/me.2003-0089] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously demonstrated that distinct facilitative glucose transporter isoforms display differential sorting in polarized epithelial cells. In Madin-Darby canine kidney (MDCK) cells, glucose transporter 1 and 2 (GLUT1 and GLUT2) are localized to the basolateral cell surface whereas GLUTs 3 and 5 are targeted to the apical membrane. To explore the molecular mechanisms underlying this asymmetric distribution, we analyzed the targeting of chimeric glucose transporter proteins in MDCK cells. Replacement of the carboxy-terminal cytosolic tail of GLUT1, GLUT2, or GLUT4 with that from GLUT3 resulted in apical targeting. Conversely, a GLUT3 chimera containing the cytosolic carboxy terminus of GLUT2 was sorted to the basolateral membrane. These findings are not attributable to the presence of a basolateral signal in the tails of GLUTs 1, 2, and 4 because the basolateral targeting of GLUT1 was retained in a GLUT1 chimera containing the carboxy terminus of GLUT5. In addition, we were unable to demonstrate the presence of an autonomous basolateral sorting signal in the GLUT1 tail using the low-density lipoprotein receptor as a reporter. By examining the targeting of a series of more defined GLUT1/3 chimeras, we found evidence of an apical targeting signal involving residues 473-484 (DRSGKDGVMEMN) in the carboxy tail. We conclude that the targeting of GLUT3 to the apical cell surface in MDCK cells is regulated by a unique cytosolic sorting motif.
Collapse
Affiliation(s)
- Kouichi Inukai
- Fourth Department of Internal Medicine, Saitama Medical School, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Heather West Greenlee M, Uemura E, Carpenter SL, Doyle RT, Buss JE. Glucose uptake in PC12 cells: GLUT3 vesicle trafficking and fusion as revealed with a novel GLUT3-GFP fusion protein. J Neurosci Res 2003; 73:518-25. [PMID: 12898536 DOI: 10.1002/jnr.10684] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The distribution of glucose transporters at the cell surface has a major impact on cellular glucose uptake. In muscle cells and adipocytes, this distribution is under the control of insulin; however, neuronal glucose uptake is not acutely regulated by insulin. Factors that affect the translocation of the neuronal glucose transporter isoform GLUT3 vesicles to and their fusion with the plasma membrane are not well understood. We report that GLUT3 in PC12 cells colocalizes with SNARE complex proteins SNAP-25 and syntaxin 1, suggesting that fusion of GLUT3-containing vesicles with the plasma membrane is mediated by these proteins. In addition, it seems that GLUT3 vesicle fusion is regulated, as depolarization increases GLUT3 insertion into the plasma membrane. To study the dynamics of GLUT3 vesicle trafficking, we have created a GLUT3-GFP fusion protein that is easily expressed in PC12 cells. Trafficking of GLUT3-GFP seems normal, as 1). its distribution is similar to endogenous GLUT3, 2). GLUT3-GFP containing vesicles fuse with the plasma membrane evidenced by labeling of the fusion protein with an antibody directed against the exofacial epitope of GLUT3, and 3). glucose uptake is similar to PC12 cells not transfected with GLUT3 fusion protein. These studies are the first to examine GLUT3 trafficking and fusion in PC12 cells, and establish a model system to study regulation of the neuronal glucose transporter.
Collapse
|
20
|
Patel JR, Brewer GJ. Age-related changes in neuronal glucose uptake in response to glutamate and beta-amyloid. J Neurosci Res 2003; 72:527-36. [PMID: 12704814 DOI: 10.1002/jnr.10602] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Energy supplies that may decline with age are crucial for cells to maintain ionic homeostasis and prevent neuron death. We examined baseline glucose transporter expression and rate of glucose uptake in cultured hippocampal neurons from embryonic, middle-age (12-month-old), and old (24-month-old) rats and exposed the neurons to glutamate, beta-amyloid, and mitochondrial inhibitors. Without stress, the rate of glucose uptake was similar in middle-age and old neurons, and the rate of glucose uptake in embryonic neurons was threefold greater than that in middle-age and old neurons. Glucose uptake increased in the presence of mitochondrial inhibitors (FCCP and oligomycin) for embryonic and middle-age neurons. The old neurons failed to increase glucose uptake. In the presence of glutamate, FCCP, and oligomycin, embryonic neurons showed a decrease in glucose uptake and the middle-age and old neurons showed no change in glucose uptake. Middle-age neurons took up significantly more glucose than old neurons when under mitochondrial and glutamate stress. In the presence of beta-amyloid, only embryonic neurons increased glucose uptake; middle-age and old neurons did not. Fluorescence imaging of immunoreactive glut3 in response to beta-amyloid demonstrated a 16-49% increase in glut3 immunoreactivity at the plasma membrane for the three ages. The results suggest that old neurons were not able to upregulate glucose uptake to ensure cell survival. Neuron aging does not indicate a defect in normal glut3 function; rather, our results suggest that mechanisms regulating glucose uptake under stress fail to react in time to ensure cell survival.
Collapse
Affiliation(s)
- Jigisha R Patel
- Department of Medical Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, Illinois 62794-9626, USA
| | | |
Collapse
|
21
|
Fladeby C, Skar R, Serck-Hanssen G. Distinct regulation of glucose transport and GLUT1/GLUT3 transporters by glucose deprivation and IGF-I in chromaffin cells. BIOCHIMICA ET BIOPHYSICA ACTA 2003; 1593:201-8. [PMID: 12581864 DOI: 10.1016/s0167-4889(02)00390-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Effects of prolonged metabolic (glucose deprivation) and hormonal [insulin-like growth factor I (IGF-I)] challenge on regulation of glucose transporter (GLUT) expression, glucose transport rate and possible signaling pathways involved were studied in the neuroendocrine chromaffin cell. The results show that bovine chromaffin cells express both GLUT1 and GLUT3. Glucose deprivation and IGF-I activation led to an elevation of GLUT1 and GLUT3 mRNA, the strongest effect being that of IGF-I on GLUT3 mRNA. Both types of stimulus increased the GLUT1 protein content in a cycloheximide (CHX)-sensitive manner, and the glucose transport rate was elevated by 3- to 4-fold after 48 h under both experimental conditions. IGF-I-induced glucose uptake was totally suppressed by CHX. In contrast, only approximately 50% of transport activation in glucose-deprived cells was sensitive to the protein synthesis inhibitor. Specific inhibitors of mTOR/FRAP and p38 MAPK each partially blocked IGF-I-stimulated glucose transport, but had no effect on transport rate in glucose-deprived cells. The results are consistent with IGF-I-activated transport being completely dependent on new GLUT protein synthesis while the enhanced transport in glucose-deprived cells was partially achieved independent of new synthesis of proteins, suggesting a mechanism relying on preexisting transporters.
Collapse
Affiliation(s)
- Cathrine Fladeby
- Department of Physiology, University of Bergen, 5009, Bergen, Norway.
| | | | | |
Collapse
|
22
|
Sakyo T, Kitagawa T. Differential localization of glucose transporter isoforms in non-polarized mammalian cells: distribution of GLUT1 but not GLUT3 to detergent-resistant membrane domains. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1567:165-75. [PMID: 12488050 DOI: 10.1016/s0005-2736(02)00613-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The hexose transporter family, which mediates a facilitated uptake in mammalian cells, consists of more than 10 members containing 12 membrane-spanning segments with a single N-glycosylation site. However, it remains unknown how these isoforms are functionally organized in the membrane domains. In this report, we describe a differential distribution of the glucose transporter isoforms GLUT1 and GLUT3 to detergent-resistant membrane domains (DRMs) in non-polarized mammalian cells. Whereas more than 80% of cellular proteins containing GLUT3 in HeLa cell lines was solubilized by a non-ionic detergent (either Triton X-100 or Lubrol WX) at 4 degrees C, GLUT1 remained insoluble together with the DRM-associated proteins, such as caveolin-1 and intestinal alkaline phosphatase (IAP). These DRM-associated proteins and the ganglioside GM1 were shown to float to the upper fractions when Triton X-100-solubilized cell extracts were centrifuged on a density gradient. In contrast, GLUT3 as well as most soluble proteins remained in the lower layers. Furthermore, perturbations of DRMs due to depletion of cholesterol by methyl-beta-cyclodextrin (m beta CD) rendered GLUT1 soluble in Triton X-100. Immunostaining patterns for these isoforms detected by confocal laser scanning microscopy in a living cell were also distinctive. These results suggest that in non-polarized mammalian cells, GLUT1 can be organized into a raft-like DRM domain but GLUT3 may distribute to fluid membrane domains. This differential distribution may occur irrespective of the N-glycosylation state or cell type.
Collapse
Affiliation(s)
- Tomoko Sakyo
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | | |
Collapse
|
23
|
Sun D, Lennernas H, Welage LS, Barnett JL, Landowski CP, Foster D, Fleisher D, Lee KD, Amidon GL. Comparison of human duodenum and Caco-2 gene expression profiles for 12,000 gene sequences tags and correlation with permeability of 26 drugs. Pharm Res 2002; 19:1400-16. [PMID: 12425456 DOI: 10.1023/a:1020483911355] [Citation(s) in RCA: 323] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE To compare gene expression profiles and drug permeability differences in Caco-2 cell culture and human duodenum. METHODS Gene expression profiles in Caco-2 cells and human duodenum were determined by GeneChip analysis. In vivo drug permeability measurements were obtained through single-pass intestinal perfusion in human subjects, and correlated with in vitro Caco-2 transport permeability. RESULTS GeneChip analysis determined that 37, 47, and 44 percent of the 12,559 gene sequences were expressed in 4-day andl6-day Caco-2 cells and human duodenum, respectively. Comparing human duodenum with Caco-2 cells, more than 1,000 sequences were determined to have at least a 5-fold difference in expression. There were 26, 38, and 44 percent of the 443 transporters, channels, and metabolizing enzymes detected in 4-day, 16-day Caco-2 cells, and human duodenum, respectively. More than 70 transporters and metabolizing enzymes exhibited at least a 3-fold difference. The overall coefficient of variability of the 10 human duodenal samples for all expressed sequences was 31% (range 3% to 294%) while that of the expressed transporters and metabolizing enzymes was 33% (range 3% to 87%). The in vivo / in vitro drug permeability measurements correlated well for passively absorbed drugs (R2 = 85%). The permeability correlation for carrier-mediated drugs showed 3- 35-fold higher in human above the correlation of passively absorbed drugs. The 2- 595-fold differences in gene expression levels between the Caco-2 cells and human duodenum correlated with the observed 3- 35-fold difference in permeability correlation between carrier-mediated drugs and passively absorbed drugs. CONCLUSIONS; Significant differences in gene expression levels in Caco-2 cells and human duodenum were observed. The observed differences of gene expression levels were consistent with observed differences in carrier mediated drug permeabilities. Gene expression profiling is a valuable new tool for investigating in vitro and in vivo permeability correlation.
Collapse
Affiliation(s)
- Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor 48109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Ardizzone TD, Lu XH, Dwyer DS. Calcium-independent inhibition of glucose transport in PC-12 and L6 cells by calcium channel antagonists. Am J Physiol Cell Physiol 2002; 283:C579-86. [PMID: 12107068 DOI: 10.1152/ajpcell.00451.2001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The goal of these studies was to determine whether different calcium channel antagonists affect glucose transport in a neuronal cell line. Rat pheochromocytoma (PC-12) cells were treated with L-, T-, and N-type calcium channel antagonists before measurement of accumulation of 2-[(3)H]deoxyglucose (2-[(3)H]DG). The L-type channel antagonists nimodipine, nifedipine, verapamil, and diltiazem all inhibited glucose transport in a dose-dependent manner (2-150 microM) with nimodipine being the most potent and diltiazem only moderately inhibiting transport. T- and N-type channel antagonists had no effect on transport. The L-type channel agonist l-BAY K 8644 also inhibited uptake of 2-[(3)H]DG. The ability of these drugs to inhibit glucose transport was significantly diminished by the presence of unlabeled 2-DG in the uptake medium. Some experiments were performed in the presence of EDTA (4 mM) or in uptake buffer without calcium. The absence of calcium in the uptake medium had no effect on inhibition of glucose transport by nimodipine or verapamil. To examine the effects of these drugs on a cell model of a peripheral tissue, we studied rat L6 muscle cells. The drugs inhibited glucose transport in L6 myoblasts in a dose-dependent manner that was independent of calcium in the uptake medium. These studies suggest that the calcium channel antagonists inhibit glucose transport in cells through mechanisms other than the antagonism of calcium channels, perhaps by acting directly on glucose transporters.
Collapse
Affiliation(s)
- Timothy D Ardizzone
- Department of Pharmacology, Louisiana State University Health Sciences Center, Shreveport, Louisiana 71130, USA
| | | | | |
Collapse
|
25
|
Piesse C, Tymms M, Garrafa E, Gouzy C, Lacasa M, Cadel S, Cohen P, Foulon T. Human aminopeptidase B (rnpep) on chromosome 1q32.2: complementary DNA, genomic structure and expression. Gene 2002; 292:129-40. [PMID: 12119107 DOI: 10.1016/s0378-1119(02)00650-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Aminopeptidase B (APB) is a Zn(2+)-metalloexopeptidase, which selectively removes Arg and/or Lys residues from the N-terminus of several peptide substrates. Several data strongly support the hypothesis that this enzyme could participate in the final stages of precursor processing mechanisms and/or in particular inflammatory processes and tumor developments. Therefore, we have cloned the complementary DNA encoding the human APB, a 658-residues protein, containing the canonical "HEXXH(X(18))E", a signature allowing its classification in the M1 family of metallopeptidases. The genomic structure of the human APB gene (rnpep; 1q32.1-q32.2) was also determined. rnpep is bracketed by pre-protein translocase of the inner mitochondrial membrane gene and ETS family transcription factor ELF3 gene. It spans more than 24 kbp and contains 11 exons ranging from 109 to 574 bp. Finally, expression of the human APB messenger RNA (mRNA) was investigated using a pre-made dot-blot. This mRNA seems to be ubiquitous although its expression level varies depending of the cells or tissues considered.
Collapse
MESH Headings
- Amino Acid Sequence
- Aminopeptidases/genetics
- Animals
- Base Sequence
- Caco-2 Cells
- Chromosomes, Human, Pair 1/genetics
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- Epoxide Hydrolases/genetics
- Female
- Gene Expression
- Genes/genetics
- HL-60 Cells
- Humans
- K562 Cells
- Male
- Mice
- Molecular Sequence Data
- Promoter Regions, Genetic/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Christophe Piesse
- Laboratoire de Biochimie des Signaux Régulateurs Cellulaires et Moléculaires, Unité Mixte de Recherche 7631 Université Pierre et Marie Curie-Centre National de la Recherche Scientifique, 96 Boulevard Raspail, F-75006 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Prapong T, Buss J, Hsu WH, Heine P, West Greenlee H, Uemura E. Amyloid beta-peptide decreases neuronal glucose uptake despite causing increase in GLUT3 mRNA transcription and GLUT3 translocation to the plasma membrane. Exp Neurol 2002; 174:253-8. [PMID: 11922666 DOI: 10.1006/exnr.2001.7861] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Amyloid beta-peptide (Abeta) has been shown to impair glucose uptake in cultured hippocampal neurons and shortens their survival time. Abeta appears to inhibit neuronal glucose uptake by activating Gs-coupled receptors and the cAMP-PKA system. In this study, Abeta inhibition of neuronal glucose uptake was studied by assaying translocation of glucose transporter isoform GLUT3, transcription of GLUT3 mRNA, and fusion of GLUT3-containing vesicles with the plasma membrane. Cultured hippocampal neurons exposed to 10 microM Abeta25-35 or Abeta1-40 for 3 or 24 h showed a significant decrease in glucose uptake. To assess the regulatory role of Abeta on neuronal glucose uptake, translocation of GLUT3 from the cytosol to the plasma membrane was studied by the plasma membrane lawn assay and transcription of GLUT3 mRNA by in situ hybridization. In spite of a decrease in glucose uptake, Abeta25-35 and Abeta1-40 (10 microM) markedly promoted GLUT3 translocation to the plasma membrane by 30 min. Abeta25-35 also up-regulated transcription of GLUT3 mRNA by 12 h. High extracellular K(+) increased immunolabeling of the exofacial (i.e., extracellular) epitope of GLUT3 at the plasma membrane and Abeta25-35 inhibited this increase. Based on these data we propose that Abeta increases translocation of GLUT3-containing vesicles, but inhibits their fusion with the plasma membrane.
Collapse
Affiliation(s)
- Teerasak Prapong
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | | | | | | | | | | |
Collapse
|
27
|
Roh C, Roduit R, Thorens B, Fried S, Kandror KV. Lipoprotein lipase and leptin are accumulated in different secretory compartments in rat adipocytes. J Biol Chem 2001; 276:35990-4. [PMID: 11451949 DOI: 10.1074/jbc.m102791200] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adipose cells produce and secrete several physiologically important proteins, such as lipoprotein lipase (LPL), leptin, adipsin, Acrp30, etc. However, secretory pathways in adipocytes have not been characterized, and vesicular carriers responsible for the accumulation and transport of secreted proteins have not been identified. We have compared the intracellular localization of two proteins secreted from adipose cells: leptin and LPL. Adipocytes accumulate large amounts of both proteins, suggesting that neither of them is targeted to the constitutive secretory pathway. By means of velocity centrifugation in sucrose gradients, equilibrium density centrifugation in iodixanol gradients, and immunofluorescence confocal microscopy, we determined that LPL and leptin were localized in different membrane structures. LPL was found mainly in the endoplasmic reticulum with a small pool being present in low density membrane vesicles that may represent a secretory compartment in adipose cells. Virtually all intracellular leptin was localized in these low density secretory vesicles. Insulin-sensitive Glut4 vesicles did not contain either LPL or leptin. Thus, secretion from adipose cells is controlled both at the exit from the endoplasmic reticulum as well as at the level of "downstream" secretory vesicles.
Collapse
Affiliation(s)
- C Roh
- Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | |
Collapse
|
28
|
Uemura E, Greenlee HW. Amyloid beta-peptide inhibits neuronal glucose uptake by preventing exocytosis. Exp Neurol 2001; 170:270-6. [PMID: 11476592 DOI: 10.1006/exnr.2001.7719] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Amyloid beta peptide (Abeta) is suspected as a contributing factor for decreased glucose utilization in the brain of Alzheimer's patients; however, little is known about the regulatory mechanism of neuronal glucose uptake and how Abeta affects such a mechanism. We report that membrane depolarization by 40 mM KCl increases both neuronal glucose uptake and immunolabeling of the exofacial epitope of glucose transporter isoform GLUT3, suggesting that fusion of GLUT3 vesicles with the plasma membrane increases glucose uptake. Abeta25-35 decreased neuronal glucose uptake and this decrease was prevented by exocytosis-enhancing compounds (40 mM KCl, 50 microM ruthenium red). Abeta25-35 also inhibited exocytosis of the fluorescent membrane dye FM1-43 at neuronal cell bodies; however, 40 mM KCl was effective in overcoming this Abeta inhibition. Furthermore, GLUT3 colocalized with SNARE (N-ethylmaleimide-sensitive factor attached protein receptor) complex proteins (SNAP-25 and Syntaxin 1), and cleavage of the v-SNARE, VAMP, reduced glucose uptake. Our findings suggest that neuronal glucose uptake is regulated by SNARE complex-dependent docking and fusion of GLUT3 vesicles with the plasma membrane and that Abeta decreases glucose uptake by inhibiting fusion of these vesicles.
Collapse
Affiliation(s)
- E Uemura
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa 50011, USA
| | | |
Collapse
|
29
|
Berg EA, Johnson RJ, Leeman SE, Boyd N, Kimerer L, Fine RE. Isolation and characterization of substance P-containing dense core vesicles from rabbit optic nerve and termini. J Neurosci Res 2000; 62:830-9. [PMID: 11107168 DOI: 10.1002/1097-4547(20001215)62:6<830::aid-jnr10>3.0.co;2-e] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In neurons, neuropeptides and other synaptic components are transported down the axon to the synapse in vesicles using molecular motors of the kinesin family. In the synapse, these neuropeptides are found in dense core vesicles (DCVs), and, following calcium-mediated exocytosis, they interact with receptors on the target cell. We have developed a rapid, large-scale technique for purifying peptide-containing DCVs from specific nuclei in the central nervous system. By using differential velocity gradient and equilibrium gradient centrifugation, neuropeptide-containing DCVs can be separated by size and density from optic nerve (ON) and its termini, the lateral geniculate nuclei and the superior colliculi. Isolated DCVs contain neuropeptides (substance P and brain-derived neurotrophic factor), synaptic vesicle (SV) membrane proteins (SV2, synaptotagmins, synaptophysin, Rab3 and synaptobrevin), SV-associated proteins (alpha-synuclein), secretory markers for DCVs previously isolated (secretogranin II), and beta-amyloid precursor protein. By using electron microscopic techniques, DCV were also visualized and shown to be immunoreactive for neuropeptides, neurotrophins, and SV membrane proteins. Because of the interesting group of physiological and potentially pathophysiological proteins associated with these vesicles; this isolation procedure, applicable to other CNS nuclei, should represent an important research tool.
Collapse
Affiliation(s)
- E A Berg
- Department of Pathology, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | | | | | |
Collapse
|
30
|
Roh C, Thoidis G, Farmer SR, Kandror KV. Identification and characterization of leptin-containing intracellular compartment in rat adipose cells. Am J Physiol Endocrinol Metab 2000; 279:E893-9. [PMID: 11001773 DOI: 10.1152/ajpendo.2000.279.4.e893] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The major leptin-containing membrane compartment was identified and characterized in rat adipose cells by means of equilibrium density and velocity sucrose gradient centrifugation. This compartment appears to be different from peptide-containing secretory granules present in neuronal, endocrine, and exocrine cells, as well as from insulin-sensitive GLUT-4-containing vesicles abundant in adipocytes. Exocytosis of both leptin- and GLUT-4-containing vesicles can be induced by insulin; however, only leptin secretion is responsive to serum stimulation. This latter effect is resistant to cycloheximide, suggesting that serum triggers the release of a stored pool of presynthesized leptin molecules. We conclude that regulated secretion of leptin and insulin-dependent translocation of GLUT-4 represent different pathways of membrane trafficking in rat adipose cells. NIH 3T3 cells ectopically expressing CAAT box enhancer binding protein-alpha and Swiss 3T3 cells expressing peroxisome proliferator-activated receptor-gamma undergo differentiation in vitro and acquire adipocyte morphology and insulin-responsive glucose uptake. Only the former cell line, however, is capable of leptin secretion. Thus different transcriptional mechanisms control the developmental onset of these two major and independent physiological functions in adipose cells.
Collapse
Affiliation(s)
- C Roh
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
31
|
Buckley KM, Melikian HE, Provoda CJ, Waring MT. Regulation of neuronal function by protein trafficking: a role for the endosomal pathway. J Physiol 2000; 525 Pt 1:11-9. [PMID: 10811720 PMCID: PMC2269916 DOI: 10.1111/j.1469-7793.2000.t01-2-00011.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Protein trafficking plays a central role in many aspects of neuronal function, from the release of neurotransmitters by exocytosis and the recycling of synaptic vesicle proteins to the regulation of receptor signalling. Synaptic function can be significantly modified on a short time scale by alterations in the levels of receptors, ion channels and transporters both pre- and postsynaptically. In many cases, these alterations appear to be mediated by acute changes in the rates at which the proteins are endocytosed from and exocytosed to the cell surface from intracellular pools. While our current understanding of the signalling mechanisms and the intracellular pathways responsible for these acute changes is still in its infancy, intriguing details are beginning to emerge from a number of systems.
Collapse
Affiliation(s)
- K M Buckley
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
32
|
Provoda CJ, Waring MT, Buckley KM. Evidence for a primary endocytic vesicle involved in synaptic vesicle biogenesis. J Biol Chem 2000; 275:7004-12. [PMID: 10702264 DOI: 10.1074/jbc.275.10.7004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulated release of neurotransmitters at synapses is mediated by the fusion of neurotransmitter-filled synaptic vesicles with the plasma membrane. Continuous synaptic activity relies on the constant recycling of synaptic vesicle proteins into newly formed synaptic vesicles. At least two different mechanisms are presumed to mediate synaptic vesicle biogenesis at the synapse as follows: direct retrieval of synaptic vesicle proteins and lipids from the plasma membrane, and indirect passage of synaptic vesicle proteins through an endosomal intermediate. We have identified a vesicle population with the characteristics of a primary endocytic vesicle responsible for the recycling of synaptic vesicle proteins through the indirect pathway. We find that synaptic vesicle proteins colocalize in this vesicle with a variety of proteins known to recycle from the plasma membrane through the endocytic pathway, including three different glucose transporters, GLUT1, GLUT3, and GLUT4, and the transferrin receptor. These vesicles differ from "classical" synaptic vesicles in their size and their generic protein content, indicating that they do not discriminate between synaptic vesicle-specific proteins and other recycling proteins. We propose that these vesicles deliver synaptic vesicle proteins that have escaped internalization by the direct pathway to endosomes, where they are sorted from other recycling proteins and packaged into synaptic vesicles.
Collapse
Affiliation(s)
- C J Provoda
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|