Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 73
1.
J Cell Biol ; 223(6)2024 Jun 03.
Article En | MEDLINE | ID: mdl-38512027

Gain-of-function mutations in the LRRK2 gene cause Parkinson's disease (PD), characterized by debilitating motor and non-motor symptoms. Increased phosphorylation of a subset of RAB GTPases by LRRK2 is implicated in PD pathogenesis. We find that increased phosphorylation of RAB3A, a cardinal synaptic vesicle precursor (SVP) protein, disrupts anterograde axonal transport of SVPs in iPSC-derived human neurons (iNeurons) expressing hyperactive LRRK2-p.R1441H. Knockout of the opposing protein phosphatase 1H (PPM1H) in iNeurons phenocopies this effect. In these models, the compartmental distribution of synaptic proteins is altered; synaptophysin and synaptobrevin-2 become sequestered in the neuronal soma with decreased delivery to presynaptic sites along the axon. We find that RAB3A phosphorylation disrupts binding to the motor adaptor MADD, potentially preventing the formation of the RAB3A-MADD-KIF1A/1Bß complex driving anterograde SVP transport. RAB3A hyperphosphorylation also disrupts interactions with RAB3GAP and RAB-GDI1. Our results reveal a mechanism by which pathogenic hyperactive LRRK2 may contribute to the altered synaptic homeostasis associated with characteristic non-motor and cognitive manifestations of PD.


Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Parkinson Disease , Synaptic Vesicles , rab3A GTP-Binding Protein , Humans , Axonal Transport , Axons , Homeostasis , Kinesins , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease/genetics , Phosphorylation , rab3A GTP-Binding Protein/genetics
2.
J Biol Chem ; 298(9): 102239, 2022 09.
Article En | MEDLINE | ID: mdl-35809645

Alpha-synuclein (a-Syn) is a presynaptic protein, the misfolding of which is associated with Parkinson's disease. Rab GTPases are small guanine nucleotide binding proteins that play key roles in vesicle trafficking and have been associated with a-Syn function and dysfunction. a-Syn is enriched on synaptic vesicles, where it has been reported to interact with GTP-bound Rab3a, a master regulator of synaptic vesicle trafficking. a-Syn is known to bind weakly to Rab8a in solution via a positively charged patch, but the physiological implications of such interactions have not been explored. Here, we investigate direct interactions between a-Syn and Rab3a in solution and on lipid membranes using NMR spectroscopy. We find that the C terminus of a-Syn interacts with Rab3a in a manner similar to its previously reported interaction with Rab8a. While weak in solution, we demonstrate that this interaction becomes stronger when the proteins are bound to a membrane surface. The Rab3a binding site for a-Syn is similar to the surface that contacts the Rab3a effector rabphilin-3A, which modulates the enzymatic activity of Rab3a. Accordingly, we show that a-Syn inhibits GTP hydrolysis by Rab3a and that inhibition is more potent on the membrane surface, suggesting that their interaction may be functionally relevant. Finally, we show that phosphorylation of a-Syn residue Ser 129, a modification associated with Parkinson's disease pathology, enhances its interactions with Rab3a and increases its ability to inhibit Rab3a GTP hydrolysis. These results represent the first observation of a functional role for synuclein-Rab interactions and for a-Syn Ser 129 phosphorylation.


Parkinson Disease , alpha-Synuclein , rab3A GTP-Binding Protein , Guanosine Triphosphate/metabolism , Humans , Lipids/chemistry , Parkinson Disease/metabolism , alpha-Synuclein/chemistry , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , rab3A GTP-Binding Protein/chemistry , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/metabolism
3.
Int J Mol Sci ; 21(15)2020 Aug 04.
Article En | MEDLINE | ID: mdl-32759795

RAB proteins (RABs) represent the largest subfamily of Ras-like small GTPases that regulate a wide variety of endosomal membrane transport pathways. Their aberrant expression has been demonstrated in various malignancies and implicated in pathogenesis. Using The Cancer Genome Atlas (TCGA) database, we analyzed the differential expression and clinicopathological association of RAB genes in pancreatic ductal adenocarcinoma (PDAC). Of the 62 RAB genes analyzed, five (RAB3A, RAB26, RAB25, RAB21, and RAB22A) exhibited statistically significant upregulation, while five (RAB6B, RAB8B, RABL2A, RABL2B, and RAB32) were downregulated in PDAC as compared to the normal pancreas. Racially disparate expression was also reported for RAB3A, RAB25, and RAB26. However, no clear trend of altered expression was observed with increasing stage and grade, age, and gender of the patients. PDAC from occasional drinkers had significantly higher expression of RAB21 compared to daily or weekly drinkers, whereas RAB25 expression was significantly higher in social drinkers, compared to occasional ones. The expression of RABL2A was significantly reduced in PDAC from diabetic patients, whereas RAB26 was significantly lower in pancreatitis patients. More importantly, a significant association of high expression of RAB21, RAB22A, and RAB25, and low expression of RAB6B, RABL2A, and RABL2B was observed with poorer survival of PC patients. Together, our study suggests potential diagnostic and prognostic significance of RABs in PDAC, warranting further investigations to define their functional and mechanistic significance.


Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Pancreatic Neoplasms/genetics , Prognosis , Adenocarcinoma/epidemiology , Adult , Aged , Aged, 80 and over , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Mitochondrial Proteins/genetics , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/pathology , Progression-Free Survival , rab GTP-Binding Proteins/genetics , rab3A GTP-Binding Protein/genetics , ras Proteins/genetics
4.
Arch Virol ; 165(2): 345-354, 2020 Feb.
Article En | MEDLINE | ID: mdl-31834525

Porcine hemagglutinating encephalomyelitis virus (PHEV) is a typical neurotropic coronavirus that mainly invades the central nervous system (CNS) in piglets and causes vomiting and wasting disease. Emerging evidence suggests that PHEV alters microRNA (miRNA) expression profiles, and miRNA has also been postulated to be involved in its pathogenesis, but the mechanisms underlying this process have not been fully explored. In this study, we found that PHEV infection upregulates miR-142a-3p RNA expression in N2a cells and in the CNS of mice. Downregulation of miR-142a-3p by an miRNA inhibitor led to a significant repression of viral proliferation, implying that it acts as a positive regulator of PHEV proliferation. Using a dual-luciferase reporter assay, miR-142a-3p was found to bind directly bound to the 3' untranslated region (3'UTR) of Rab3a mRNA and downregulate its expression. Knockdown of Rab3a expression by transfection with an miR-142a-3p mimic or Rab3a siRNA significantly increased PHEV replication in N2a cells. Conversely, the use of an miR-142a-3p inhibitor or overexpression of Rab3a resulted in a marked restriction of viral production at both the mRNA and protein level. Our data demonstrate that miR-142a-3p promotes PHEV proliferation by directly targeting Rab3a mRNA, and this provides new insights into the mechanisms of PHEV-related pathogenesis and virus-host interactions.


Betacoronavirus 1/genetics , Cell Proliferation/genetics , Coronavirus Infections/genetics , MicroRNAs/genetics , Swine/virology , rab3A GTP-Binding Protein/genetics , 3' Untranslated Regions/genetics , Animals , Cell Line , Cell Line, Tumor , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Down-Regulation/genetics , HEK293 Cells , Humans , Mice , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Up-Regulation/genetics
5.
J Cell Sci ; 131(23)2018 11 29.
Article En | MEDLINE | ID: mdl-30404828

α-Synuclein is associated with Parkinson's disease, and is mainly localized in presynaptic terminals and regulates exocytosis, but its physiological roles remain controversial. Here, we studied the effects of soluble and aggregated α-synuclein on exocytosis, and explored the molecular mechanism by which α-synuclein interacts with regulatory proteins, including Rab3A, Munc13-1 (also known as Unc13a) and Munc18-1 (also known as STXBP1), in order to regulate exocytosis. Through fluorescence recovery after photobleaching experiments, overexpressed α-synuclein in PC12 cells was found to be in a monomeric form, which promotes exocytosis. In contrast, aggregated α-synuclein induced by lactacystin treatment inhibits exocytosis. Our results show that α-synuclein is involved in vesicle priming and fusion. α-Synuclein and phorbol 12-myristate 13-acetate (PMA), which is known to enhance vesicle priming mediated by Rab3A, Munc13-1 and Munc18-1, act on the same population of vesicles, but regulate priming independently. Furthermore, the results show a novel effects of α-synuclein on mobilizing Ca2+ release from thapsigargin-sensitive Ca2+ pools to enhance the ATP-induced [Ca2+]i increase, which enhances vesicle fusion. Our results provide a detailed understanding of the action of α-synuclein during the final steps of exocytosis.


Calcium/metabolism , Exocytosis/physiology , Thapsigargin/pharmacology , alpha-Synuclein/metabolism , Animals , Membrane Fusion/physiology , PC12 Cells , Rats , Thapsigargin/metabolism , Transfection , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/metabolism
6.
Cell Death Dis ; 9(10): 970, 2018 09 20.
Article En | MEDLINE | ID: mdl-30237463

Rab3A is a small Ras-like GTPase critical for membrane traffic. Although the functions of Rab3A have been reported in several cancers, the roles of Rab3A in hepatocellular carcinoma (HCC) have never been determined. To investigate the potential roles of Rab3A in HCC progression, we first determined Rab3A levels in HCC tissues and observed upregulated mRNA and protein levels of Rab3A in most tumor tissues. However, in vitro data showed that decreasing Rab3A in most HCC cell lines conferred no significant effects and overexpressing Rab3A in PLC/PRF/5 cells even inhibited migration and invasion. Meanwhile, the upregulation of Rab3A in HCC patients did not correlate with metastasis or overall survival of HCC patients. These contradict data suggested that Rab3A might act as metastatic suppressor and its effects might be attenuated in most HCC cells. Further experiments revealed that O-GlcNAcylation on Rab3A was key for attenuating Rab3A-mediated effects by regulating its GTP-binding activity, and verified the effects of Rab3A and its aberrant O-GlcNAcylation on HCC metastasis in vitro and in vivo. We also found that Rab3A and its O-GlcNAcylation had opposite roles in mitochondria oxidative phosphorylation (mtOXPHOS), and their functions on HCC metastasis were partially depended on their effects on metabolic reprogramming.


Acetylglucosamine/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , rab3A GTP-Binding Protein/metabolism , Adult , Animals , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Hep G2 Cells , Humans , Hydrogen Peroxide/metabolism , Immunohistochemistry , Immunoprecipitation , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Phosphorylation , Oxygen Consumption/physiology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/physiology , Superoxides/metabolism , rab3A GTP-Binding Protein/genetics
7.
Sci Rep ; 8(1): 10957, 2018 Jul 19.
Article En | MEDLINE | ID: mdl-30026496

Functional electrical stimulation (FES) is rapidly gaining traction as a therapeutic tool for mediating the repair and recovery of the injured central nervous system (CNS). However, the underlying mechanisms and impact of these stimulation paradigms at a molecular, cellular and network level remain largely unknown. In this study, we used embryonic stem cell (ESC)-derived neuron and glial co-cultures to investigate network maturation following acute administration of L-glutamate, which is a known mediator of excitotoxicity following CNS injury. We then modulated network maturation using chronic low frequency stimulation (LFS) and direct current stimulation (DCS) protocols. We demonstrated that L-glutamate impaired the rate of maturation of ESC-derived neurons and glia immediately and over a week following acute treatment. The administration of chronic LFS and DCS protocols individually following L-glutamate infusion significantly promoted the excitability of neurons as well as network synchrony, while the combination of LFS/DCS did not. qRT-PCR analysis revealed that LFS and DCS alone significantly up-regulated the expression of excitability and plasticity-related transcripts encoding N-methyl-D-aspartate (NMDA) receptor subunit (NR2A), brain-derived neurotrophic factor (BDNF) and Ras-related protein (RAB3A). In contrast, the simultaneous administration of LFS/DCS down-regulated BDNF and RAB3A expression. Our results demonstrate that LFS and DCS stimulation can modulate network maturation excitability and synchrony following the acute administration of an inhibitory dose of L-glutamate, and upregulate NR2A, BDNF and RAB3A gene expression. Our study also provides a novel framework for investigating the effects of electrical stimulation on neuronal responses and network formation and repair after traumatic brain injury.


Electric Stimulation/methods , Glutamic Acid/pharmacology , Neuroglia/cytology , Neuronal Plasticity , Neurons/cytology , Animals , Brain-Derived Neurotrophic Factor/genetics , Cells, Cultured , Coculture Techniques/methods , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Neuroglia/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Up-Regulation , rab3A GTP-Binding Protein/genetics
8.
J Cell Biochem ; 118(11): 3696-3705, 2017 11.
Article En | MEDLINE | ID: mdl-28370453

Rab3 and synaptotagmin have been suggested to play important roles in the regulation of neurotransmitter release and, however, the molecular mechanism has not been completely clear. Here, we studied the effects of Rab3A and synaptotagmin I (Syt I) on dopamine release using PC12 cells as a model system. Rab3A was demonstrated to have effects on both Ca2+ -independent and Ca2+ -dependent dopamine releases from the PC12 cells. Application of Rab3A (up to 2500 nM) gradually decreased the amount of Ca2+ -dependently released dopamine, indicating that Rab3A is a negative modulator that was further supported by the increase in dopamine release caused by Rab3A knockdown. Syt I knockdown weakened the Ca2+ -dependent dopamine release, suggesting that Syt I plays a positive regulatory role in the cellular process. Treatment of the Syt I-knocked down PC12 cells with Rab3A further decreased Ca2+ -dependent dopamine release and, however, the decrease magnitude was significantly reduced compared with that before Syt I knockdown, thus for the first time demonstrating that the inhibitory effect of Rab3A on Ca2+ -dependent dopamine release involves the interaction with Syt I. This work has shed new light on the molecular mechanism for Rab3 and synaptotamin regulation of neurotransmitter release. J. Cell. Biochem. 118: 3696-3705, 2017. © 2017 Wiley Periodicals, Inc.


Calcium/metabolism , Dopamine/metabolism , Neurotransmitter Agents/metabolism , Synaptotagmin I/metabolism , rab3A GTP-Binding Protein/metabolism , Animals , Gene Knockdown Techniques , PC12 Cells , Rats , Synaptotagmin I/genetics , rab3A GTP-Binding Protein/antagonists & inhibitors , rab3A GTP-Binding Protein/genetics
9.
J Biol Chem ; 291(44): 23101-23111, 2016 10 28.
Article En | MEDLINE | ID: mdl-27613869

At the final stage of exocytotis, a fusion pore opens between the plasma and a secretory vesicle membranes; typically, when the pore dilates the vesicle releases its cargo. Sperm contain a large dense-core secretory granule (the acrosome) whose contents are secreted by regulated exocytosis at fertilization. Minutes after the arrival of the triggering signal, the acrosomal and plasma membranes dock at multiple sites and fusion pores open at the contact points. It is believed that immediately afterward, fusion pores dilate spontaneously. Rab3A is an essential component of human sperm exocytotic machinery. Yet, recombinant, persistently active Rab3A halts calcium-triggered secretion when introduced after docking into streptolysin O-permeabilized cells; so does a Rab3A-22A chimera. Here, we applied functional assays, electron and confocal microscopy to show that the secretion blockage is due to the stabilization of open fusion pores. Other novel findings are that sperm SNAREs engage in α-SNAP/NSF-sensitive complexes at a post-fusion stage. Complexes are disentangled by these chaperons to achieve vesiculation and acrosomal contents release. Thus, post-fusion regulation of the pores determines their expansion and the success of the acrosome reaction.


Exocytosis , Spermatozoa/metabolism , rab GTP-Binding Proteins/metabolism , rab3A GTP-Binding Protein/metabolism , Acrosome/metabolism , Calcium/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Humans , Male , rab GTP-Binding Proteins/genetics , rab3A GTP-Binding Protein/genetics
10.
Histochem Cell Biol ; 145(6): 647-57, 2016 Jun.
Article En | MEDLINE | ID: mdl-26791531

Rab family members play important roles in membrane trafficking, cell growth, and differentiation. Almost all components of the cell endomembrane system, the nucleus, and the plasma membrane are closely related to RAB proteins. In this study, we investigated the distribution and functions of three members of the Rab family, Rab3A, Rab27A, and Rab35, in mouse oocyte meiotic maturation and activation. The three Rab family members showed different localization patterns in oocytes. Microinjection of siRNA, antibody injection, or inhibitor treatment showed that (1) Rab3A regulates peripheral spindle and cortical granule (CG) migration, polarity establishment, and asymmetric division; (2) Rab27A regulates CG exocytosis following MII-stage oocyte activation; and (3) Rab35 plays an important role in spindle organization and morphology maintenance, and thus meiotic nuclear maturation. These results show that Rab proteins play important roles in mouse oocyte meiotic maturation and activation and that different members exert different distinct functions.


Meiosis , Oocytes/cytology , Oocytes/metabolism , rab GTP-Binding Proteins/metabolism , rab3A GTP-Binding Protein/metabolism , Animals , Cells, Cultured , Fluorescent Antibody Technique , Mice , Mice, Inbred ICR , rab GTP-Binding Proteins/analysis , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins , rab3A GTP-Binding Protein/analysis , rab3A GTP-Binding Protein/genetics
11.
J Endocrinol ; 223(3): 267-75, 2014 Dec.
Article En | MEDLINE | ID: mdl-25274988

The Src homology-2 domain containing protein B (SHB) has previously been shown to function as a pleiotropic adapter protein, conveying signals from receptor tyrosine kinases to intracellular signaling intermediates. The overexpression of Shb in ß-cells promotes ß-cell proliferation by increased insulin receptor substrate (IRS) and focal adhesion kinase (FAK) activity, whereas Shb deficiency causes moderate glucose intolerance and impaired first-peak insulin secretion. Using an array of techniques, including live-cell imaging, patch-clamping, immunoblotting, and semi-quantitative PCR, we presently investigated the causes of the abnormal insulin secretory characteristics in Shb-knockout mice. Shb-knockout islets displayed an abnormal signaling signature with increased activities of FAK, IRS, and AKT. ß-catenin protein expression was elevated and it showed increased nuclear localization. However, there were no major alterations in the gene expression of various proteins involved in the ß-cell secretory machinery. Nor was Shb deficiency associated with changes in glucose-induced ATP generation or cytoplasmic Ca(2+) handling. In contrast, the glucose-induced rise in cAMP, known to be important for the insulin secretory response, was delayed in the Shb-knockout compared with WT control. Inhibition of FAK increased the submembrane cAMP concentration, implicating FAK activity in the regulation of insulin exocytosis. In conclusion, Shb deficiency causes a chronic increase in ß-cell FAK activity that perturbs the normal insulin secretory characteristics of ß-cells, suggesting multi-faceted effects of FAK on insulin secretion depending on the mechanism of FAK activation.


Focal Adhesion Protein-Tyrosine Kinases/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Proto-Oncogene Proteins/deficiency , Adenosine Triphosphate/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cyclic AMP/metabolism , Exocytosis/genetics , Gene Expression , Glucose/pharmacology , Immunoblotting , Insulin Receptor Substrate Proteins/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Membrane Potentials , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence/methods , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , beta Catenin/metabolism , rab3A GTP-Binding Protein/genetics
12.
Mol Biol Rep ; 41(9): 5903-11, 2014 Sep.
Article En | MEDLINE | ID: mdl-24965146

The Rab protein family is composed of small GTP-binding proteins involved in intracellular vesicle trafficking. In particular, Rab3a which is one of four Rab3 proteins (a, b, c, and d isoforms) is associated with synaptic vesicle trafficking in normal brain. However, despite the elevated level of Rab3a in tumors, its role remains unclear. Here we report a tumorigenic role of Rab3a in brain tumors. Elevated level of Rab3a expression in human was confirmed in both glioma cell lines and glioblastoma multiforme patient specimens. Ectopic Rab3a expression in glioma cell lines and primary astrocytes promoted cell proliferation by increasing cyclin D1 expression, induced resistance to anti-cancer drug and irradiation, and accelerated foci formation in soft agar and tumor formation in nude mice. The overexpression of Rab3a augmented the tumorsphere-forming ability of glioma cells and p53(-/-) astrocytes and increased expression levels of various stem cell markers. Taken together, our results indicate that Rab3a is a novel oncogene involved in glioma initiation and progression.


Brain Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , rab3A GTP-Binding Protein/metabolism , Animals , Astrocytes/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Progression , Drug Resistance, Neoplasm/genetics , Glioblastoma/genetics , Humans , Mice , Mice, Knockout , Mice, Nude , rab3A GTP-Binding Protein/genetics
13.
Mol Biol Rep ; 41(6): 3951-9, 2014 Jun.
Article En | MEDLINE | ID: mdl-24652202

Rab3A is expressed predominantly in brain and synaptic vesicles. Rab3A is involved specifically in tethering and docking of synaptic vesicles prior to fusion which is a critical step in regulated release of neurotransmitters. The precise function of Rab3A is still not known. However, up-regulation of Rab3A has been reported in malignant neuroendocrine and breast cancer cells. In the present study, the structure of Rab3A protein was generated using MODELLER 9v8 software. The modeled protein structure was validated and subjected to molecular docking analyses. Docking with GTP was carried out on the binding site of Rab3A using GOLD software. The Rab3A-GTP complex has best GOLD fitness value of 77.73. Ligplot shows hydrogen bondings (S16, S17, V18, G19, K20, T21, S22, S31, T33, A35, S38, T39 and G65) and hydrophobic interacting residues (F25, F32, P34, F36, V37, D62 and A64) with the GTP ligands in the binding site of Rab3A protein. Here, the ligand molecules of NCI diversity set II from the ZINC database against the active site of the Rab3A protein were screened. For this purpose, the incremental construction algorithm of GLIDE and the genetic algorithm of GOLD were used. Docking results were analyzed for top ranking compounds using a consensus scoring function of X-Score to calculate the binding affinity and Ligplot was used to measure protein-ligand interactions. Five compounds which possess good inhibitory activity and may act as potential high affinity inhibitors against Rab3A active site were identified. The top ranking molecule (ZINC13152284) has a Glide score of -6.65 kcal/mol, X-Score of -3.02 kcal/mol and GOLD score of 64.54 with 03 hydrogen bonds and 09 hydrophobic contacts. This compound is thus a good starting point for further development of strong inhibitors.


Carcinogenesis/chemistry , Molecular Docking Simulation , rab3A GTP-Binding Protein/chemistry , Binding Sites , Catalytic Domain , Humans , Ligands , Molecular Conformation , Small Molecule Libraries/chemistry , rab3A GTP-Binding Protein/antagonists & inhibitors , rab3A GTP-Binding Protein/genetics
14.
J Neurosci ; 33(46): 18319-30, 2013 Nov 13.
Article En | MEDLINE | ID: mdl-24227741

Some forms of idiopathic epilepsy in animals and humans are associated with deficiency of synapsin, a phosphoprotein that reversibly associates with synaptic vesicles. We have previously shown that the epileptic phenotype seen in synapsin II knock-out mice (SynII(-)) can be rescued by the genetic deletion of the Rab3a protein. Here we have examined the cellular basis for this rescue using whole-cell recordings from CA1 hippocampal pyramidal cells in brain slices. We find that SynII(-) neurons have increased spontaneous activity and a reduced threshold for the induction of epileptiform activity by 4-aminopyridine (4-AP). Using selective recordings of glutamatergic and GABAergic activity we show that in wild-type neurons low concentrations of 4-AP facilitate glutamatergic and GABAergic transmission in a balanced way, whereas in SynII(-) neurons this balance is shifted toward excitation. This imbalance reflects a deficit in inhibitory synaptic transmission that appears to be secondary to reduced Ca(2+) sensitivity in SynII(-) neurons. This suggestion is supported by our finding that synaptic and epileptiform activity at SynII(-) and wild-type synapses is similar when GABAergic transmission is blocked. Deletion of Rab3a results in glutamatergic synapses that have a compromised responsiveness to either low 4-AP concentrations or elevated extracellular Ca(2+). These changes mitigate the overexcitable phenotype observed in SynII(-) neurons. Thus, Rab3a deletion appears to restore the excitatory/inhibitory imbalance observed in SynII(-) hippocampal slices indirectly, not by correcting the deficit in GABAergic synaptic transmission but rather by impairing excitatory glutamatergic synaptic transmission.


CA1 Region, Hippocampal/metabolism , Epilepsy/metabolism , Synapses/metabolism , Synapsins/deficiency , rab3A GTP-Binding Protein/deficiency , Animals , Epilepsy/genetics , Epilepsy/physiopathology , Excitatory Postsynaptic Potentials/physiology , Female , Male , Mice , Mice, Knockout , Organ Culture Techniques , Synapses/genetics , Synapsins/genetics , rab3A GTP-Binding Protein/genetics
15.
PLoS One ; 8(10): e78883, 2013.
Article En | MEDLINE | ID: mdl-24205339

Rab3a is a small GTPase of the Rab3 subfamily that acts during late stages of Ca²âº-regulated exocytosis. Previous functional analysis in pituitary melanotrophs described Rab3a as a positive regulator of Ca²âº-dependent exocytosis. However, the precise role of the Rab3a isoform on the kinetics and intracellular [Ca²âº] sensitivity of regulated exocytosis, which may affect the availability of two major peptide hormones, α-melanocyte stimulating hormone (α-MSH) and ß-endorphin in plasma, remain elusive. We employed Rab3a knock-out mice (Rab3a KO) to explore the secretory phenotype in melanotrophs from fresh pituitary tissue slices. High resolution capacitance measurements showed that Rab3a KO melanotrophs possessed impaired Ca²âº-triggered secretory activity as compared to wild-type cells. The hampered secretion was associated with the absence of cAMP-guanine exchange factor II/ Epac2-dependent secretory component. This component has been attributed to high Ca²âº-sensitive release-ready vesicles as determined by slow photo-release of caged Ca²âº. Radioimmunoassay revealed that α-MSH, but not ß-endorphin, was elevated in the plasma of Rab3a KO mice, indicating increased constitutive exocytosis of α-MSH. Increased constitutive secretion of α-MSH from incubated tissue slices was associated with reduced α-MSH cellular content in Rab3a-deficient pituitary cells. Viral re-expression of the Rab3a protein in vitro rescued the secretory phenotype of melanotrophs from Rab3a KO mice. In conclusion, we suggest that Rab3a deficiency promotes constitutive secretion and underlies selective impairment of Ca²âº-dependent release of α-MSH.


Calcium/metabolism , Exocytosis , Melanotrophs/cytology , Secretory Vesicles/metabolism , alpha-MSH/metabolism , rab3A GTP-Binding Protein/metabolism , Animals , Cyclic AMP/pharmacology , Exocytosis/drug effects , Gene Knockout Techniques , Melanotrophs/drug effects , Mice , Mice, Knockout , Secretory Vesicles/drug effects , rab3A GTP-Binding Protein/deficiency , rab3A GTP-Binding Protein/genetics
16.
J Biol Chem ; 288(45): 32466-32474, 2013 Nov 08.
Article En | MEDLINE | ID: mdl-24072714

Small G-proteins of the Ras superfamily control the temporal and spatial coordination of intracellular signaling networks by acting as molecular on/off switches. Guanine nucleotide exchange factors (GEFs) regulate the activation of these G-proteins through catalytic replacement of GDP by GTP. During nucleotide exchange, three distinct substrate·enzyme complexes occur: a ternary complex with GDP at the start of the reaction (G-protein·GEF·GDP), an intermediary nucleotide-free binary complex (G-protein·GEF), and a ternary GTP complex after productive G-protein activation (G-protein·GEF·GTP). Here, we show structural snapshots of the full nucleotide exchange reaction sequence together with the G-protein substrates and products using Rabin8/GRAB (GEF) and Rab8 (G-protein) as a model system. Together with a thorough enzymatic characterization, our data provide a detailed view into the mechanism of Rabin8/GRAB-mediated nucleotide exchange.


Guanosine Triphosphate/chemistry , Multienzyme Complexes/chemistry , Protein Serine-Threonine Kinases/chemistry , rab GTP-Binding Proteins/chemistry , rab3A GTP-Binding Protein/chemistry , Catalysis , Germinal Center Kinases , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , Humans , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/metabolism
17.
PLoS One ; 8(6): e68255, 2013.
Article En | MEDLINE | ID: mdl-23826383

Rab37 belongs to a subclass of Rab GTPases regulating exocytosis, including also Rab3a and Rab27a. Proteomic studies indicate that Rab37 is associated with insulin-containing large dense core granules of pancreatic ß-cells. In agreement with these observations, we detected Rab37 in extracts of ß-cell lines and human pancreatic islets and confirmed by confocal microscopy the localization of the GTPase on insulin-containing secretory granules. We found that, as is the case for Rab3a and Rab27a, reduction of Rab37 levels by RNA interference leads to impairment in glucose-induced insulin secretion and to a decrease in the number of granules in close apposition to the plasma membrane. Pull-down experiments revealed that, despite similar functional effects, Rab37 does not interact with known Rab3a or Rab27a effectors and is likely to operate through a different mechanism. Exposure of insulin-secreting cells to proinflammatory cytokines, fatty acids or oxidized low-density lipoproteins, mimicking physiopathological conditions that favor the development of diabetes, resulted in a decrease in Rab37 expression. Our data identify Rab37 as an additional component of the machinery governing exocytosis of ß-cells and suggest that impaired expression of this GTPase may contribute to defective insulin release in pre-diabetic and diabetic conditions.


Exocytosis/physiology , Insulin/metabolism , rab GTP-Binding Proteins/metabolism , Cell Line , Fluorescent Antibody Technique , Gene Knockdown Techniques , Glucose/administration & dosage , Glucose/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Human Growth Hormone/metabolism , Humans , Immunoblotting , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/enzymology , Islets of Langerhans/metabolism , Microscopy, Fluorescence , RNA Interference , Real-Time Polymerase Chain Reaction , Transfection , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/metabolism
18.
PLoS One ; 8(4): e62893, 2013.
Article En | MEDLINE | ID: mdl-23658655

The calyx of Held, a large glutamatergic terminal in the mammalian auditory brainstem has been extensively employed to study presynaptic structure and function in the central nervous system. Nevertheless, the nanoarchitecture of presynaptic proteins and subcellular components in the calyx terminal and its relation to functional properties of synaptic transmission is only poorly understood. Here, we use stimulated emission depletion (STED) nanoscopy of calyces in thin sections of aldehyde-fixed rat brain tissue to visualize immuno-labeled synaptic proteins including VGluT1, synaptophysin, Rab3A and synapsin with a lateral resolution of approximately 40 nm. Excitation multiplexing of suitable fluorescent dyes deciphered the spatial arrangement of the presynaptic phospho-protein synapsin relative to synaptic vesicles labeled with anti-VGluT1. Both predominantly occupied the same focal volume, yet may exist in exclusive domains containing either VGluT1 or synapsin immunoreactivity. While the latter have been observed with diffraction-limited fluorescence microscopy, STED microscopy for the first time revealed VGluT1-positive domains lacking synapsins. This observation supports the hypothesis that molecularly and structurally distinct synaptic vesicle pools operate in presynaptic nerve terminals.


Auditory Cortex/ultrastructure , Presynaptic Terminals/ultrastructure , Synaptic Transmission/physiology , Synaptic Vesicles/ultrastructure , Animals , Auditory Cortex/metabolism , Fixatives , Fluorescent Dyes , Gene Expression , Microscopy, Fluorescence/methods , Microtomy , Phosphorylation , Presynaptic Terminals/metabolism , Rats , Rats, Sprague-Dawley , Synapsins/genetics , Synapsins/metabolism , Synaptic Vesicles/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/metabolism
19.
Gen Comp Endocrinol ; 185: 67-79, 2013 May 01.
Article En | MEDLINE | ID: mdl-23399968

In this work we have compared the ultrastructural characteristics of major pancreatic endocrine cells, pituitary melanotrophs and adrenal chromaffin cells in the normal mouse strain (wild type, WT) and mice with a known secretory deficit, the Rab3a knockout strain (Rab3a KO). For this purpose, pancreata, pituitary glands and adrenal glands from the Rab3a KO and from the WT mice were analysed, using conventional transmission electron microscopy (TEM). In order to assess the significance of the presence of Rab3a proteins in the relevant cells, we focused primarily on their secretory vesicle morphology and distribution. Our results showed a comparable general morphology in Rab3a KO and WT in all assessed endocrine cell types. In all studied cell types, the distribution of secretory granules along the plasma membrane (number of docked and almost-docked vesicles) was comparable between Rab3a KO and WT mice. Specific differences were found in the diameters of their secretory vesicles, diameters of their electron-dense cores and the presence of autophagic structures in the cells of Rab3A KO mice only. Occasionally, individual electron-dense round vesicles were present inside autophagosome-like structures; these were possibly secretory vesicles or their remnants. The differences found in the diameters of the secretory vesicles confirm the key role of Rab3a proteins in controlling the balance between secretory vesicle biogenesis and degradation, and suggest that the ablation of this protein probably changes the nature of the reservoir of secretory vesicles available for regulated exocytosis.


Chromaffin Cells/ultrastructure , Melanotrophs/ultrastructure , Pancreas/ultrastructure , Secretory Vesicles/ultrastructure , rab3A GTP-Binding Protein/deficiency , Adrenal Glands/ultrastructure , Animals , Exocytosis , Male , Mice , Mice, Knockout , Pituitary Gland/ultrastructure , Secretory Vesicles/physiology , rab3A GTP-Binding Protein/genetics
20.
J Neurosci ; 32(20): 6931-6, 2012 May 16.
Article En | MEDLINE | ID: mdl-22593061

Rab3A is a synaptic vesicle-associated protein found throughout the nervous system, but its precise function is unknown. Genetic knock-out studies show that Rab3A is not necessary for vesicular release or replenishment at conventional synapses in the brain. Here we explore the function of Rab3A at ribbon synapses in the retina of the tiger salamander (Ambystoma tigrinum). Fluorescently labeled Rab3A, delivered into rods and cones through a patch pipette, binds to and dissociates from synaptic ribbons. Experiments using nonphosphorylatable GDP analogs and a GTPase-deficient Rab3A mutant indicate that ribbon binding and dissociation are governed by a GTP hydrolysis cycle. Paired recordings from presynaptic photoreceptors and postsynaptic OFF-bipolar cells show that the Rab3A mutant blocks synaptic release in an activity-dependent manner, with more frequent stimulation leading to more rapid block. The frequency dependence of block by exogenous Rab3A suggests that it acts competitively with synaptic vesicles to interfere with their resupply to release sites. Together, these findings suggest a crucial role of Rab3A in delivering vesicles to Ca²âº-dependent release sites at ribbon synapses.


Photoreceptor Cells, Vertebrate/physiology , Synapses/metabolism , Synaptic Vesicles/metabolism , rab3A GTP-Binding Protein/metabolism , Ambystoma , Animals , Female , GTP Phosphohydrolases/genetics , Male , Mutation , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Retina/drug effects , Retina/metabolism , Retinal Bipolar Cells/physiology , rab3A GTP-Binding Protein/genetics , rab3A GTP-Binding Protein/pharmacology
...