Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(10)2023 09 28.
Article in English | MEDLINE | ID: mdl-37892145

ABSTRACT

Retinal neurons that form ribbon-style synapses operate over a wide dynamic range, continuously relaying visual information to their downstream targets. The remarkable signaling abilities of these neurons are supported by specialized presynaptic machinery, one component of which is syntaxin3B. Syntaxin3B is an essential t-SNARE protein of photoreceptors and bipolar cells that is required for neurotransmitter release. It has a light-regulated phosphorylation site in its N-terminal domain at T14 that has been proposed to modulate membrane fusion. However, a direct test of the latter has been lacking. Using a well-controlled in vitro fusion assay, we found that a phosphomimetic T14 syntaxin3B mutation leads to a small but significant enhancement of SNARE-mediated membrane fusion following the formation of the t-SNARE complex. While the addition of Munc18a had only a minimal effect on membrane fusion mediated by SNARE complexes containing wild-type syntaxin3B, a more significant enhancement was observed in the presence of Munc18a when the SNARE complexes contained a syntaxin3B T14 phosphomimetic mutant. Finally, we showed that the retinal-specific complexins (Cpx III and Cpx IV) inhibited membrane fusion mediated by syntaxin3B-containing SNARE complexes in a dose-dependent manner. Collectively, our results establish that membrane fusion mediated by syntaxin3B-containing SNARE complexes is regulated by the T14 residue of syntaxin3B, Munc18a, and Cpxs III and IV.


Subject(s)
Membrane Fusion , Synapses , Membrane Fusion/physiology , Synapses/metabolism , Synaptic Transmission/genetics , Retina/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Protein Binding
2.
Article in English | MEDLINE | ID: mdl-37609371

ABSTRACT

Biallelic loss-of-function mutations in the syntaxin 3 gene have been linked to a severe retinal dystrophy in humans that presents in early childhood. In mouse models, biallelic inactivation of the syntaxin 3 gene in photoreceptors rapidly leads to their death. What is not known is whether a monoallelic syntaxin 3 loss-of-function mutation might cause photoreceptor loss with advancing age. To address this question, we compared the outer nuclear layer of older adult mice (≈ 20 months of age) that were heterozygous for syntaxin 3 with those of similarly-aged control mice. We found that the photoreceptor layer maintains its thickness in mice that are heterozygous for syntaxin 3 relative to controls and that photoreceptor somatic counts are comparable. In addition, dendritic sprouting of the rod bipolar cell dendrites into the outer nuclear layer, which occurs following the loss of functional rod targets, was similar between genotypes. Thus, syntaxin 3 appears to be haplosufficient for photoreceptor survival, even with advancing age.

3.
Hum Genet ; 140(8): 1143-1156, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33974130

ABSTRACT

Biallelic STX3 variants were previously reported in five individuals with the severe congenital enteropathy, microvillus inclusion disease (MVID). Here, we provide a significant extension of the phenotypic spectrum caused by STX3 variants. We report ten individuals of diverse geographic origin with biallelic STX3 loss-of-function variants, identified through exome sequencing, single-nucleotide polymorphism array-based homozygosity mapping, and international collaboration. The evaluated individuals all presented with MVID. Eight individuals also displayed early-onset severe retinal dystrophy, i.e., syndromic-intestinal and retinal-disease. These individuals harbored STX3 variants that affected both the retinal and intestinal STX3 transcripts, whereas STX3 variants affected only the intestinal transcript in individuals with solitary MVID. That STX3 is essential for retinal photoreceptor survival was confirmed by the creation of a rod photoreceptor-specific STX3 knockout mouse model which revealed a time-dependent reduction in the number of rod photoreceptors, thinning of the outer nuclear layer, and the eventual loss of both rod and cone photoreceptors. Together, our results provide a link between STX3 loss-of-function variants and a human retinal dystrophy. Depending on the genomic site of a human loss-of-function STX3 variant, it can cause MVID, the novel intestinal-retinal syndrome reported here or, hypothetically, an isolated retinal dystrophy.


Subject(s)
Eye Diseases, Hereditary/genetics , Intestinal Mucosa/metabolism , Malabsorption Syndromes/genetics , Microvilli/pathology , Mucolipidoses/genetics , Polymorphism, Single Nucleotide , Qa-SNARE Proteins/genetics , Retinal Cone Photoreceptor Cells/metabolism , Retinal Dystrophies/genetics , Aged , Aged, 80 and over , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Animals , Autopsy , Co-Repressor Proteins/genetics , Co-Repressor Proteins/metabolism , Eye Diseases, Hereditary/metabolism , Eye Diseases, Hereditary/pathology , Female , Gene Expression Regulation , Homozygote , Humans , Intestinal Mucosa/pathology , Malabsorption Syndromes/metabolism , Malabsorption Syndromes/pathology , Mice , Mice, Knockout , Microvilli/genetics , Microvilli/metabolism , Mucolipidoses/metabolism , Mucolipidoses/pathology , Phenotype , Qa-SNARE Proteins/deficiency , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Cone Photoreceptor Cells/pathology , Retinal Dystrophies/metabolism , Retinal Dystrophies/pathology , Sensory Rhodopsins/genetics , Sensory Rhodopsins/metabolism , Exome Sequencing
4.
J Biol Chem ; 296: 100268, 2021.
Article in English | MEDLINE | ID: mdl-33837726

ABSTRACT

Degranulation, a fundamental effector response from mast cells (MCs) and platelets, is an example of regulated exocytosis. This process is mediated by SNARE proteins and their regulators. We have previously shown that several of these proteins are essential for exocytosis in MCs and platelets. Here, we assessed the role of the SNARE protein SNAP23 using conditional knockout mice, in which SNAP23 was selectively deleted from either the megakaryocyte/platelet or connective tissue MC lineages. We found that removal of SNAP23 in platelets results in severe defects in degranulation of all three platelet secretory granule types, i.e., alpha, dense, and lysosomal granules. The mutation also induces thrombocytopenia, abnormal platelet morphology and activation, and reduction in the number of alpha granules. Therefore, the degranulation defect might not be secondary to an intrinsic failure of the machinery mediating regulated exocytosis in platelets. When we removed SNAP23 expression in MCs, there was a complete developmental failure in vitro and in vivo. The developmental defects in platelets and MCs and the abnormal translocation of membrane proteins to the surface of platelets indicate that SNAP23 is also involved in constitutive exocytosis in these cells. The MC conditional deletant animals lacked connective tissue MCs, but their mucosal MCs were normal and expanded in response to an antigenic stimulus. We used this mouse to show that connective tissue MCs are required and mucosal MCs are not sufficient for an anaphylactic response.


Subject(s)
Anaphylaxis/immunology , Blood Platelets/immunology , Connective Tissue/immunology , Mast Cells/immunology , Qb-SNARE Proteins/immunology , Qc-SNARE Proteins/immunology , Anaphylaxis/genetics , Anaphylaxis/pathology , Animals , Blood Platelets/pathology , Connective Tissue/pathology , Exocytosis/genetics , Exocytosis/immunology , Mast Cells/pathology , Mice , Mice, Knockout , Qb-SNARE Proteins/genetics , Qc-SNARE Proteins/genetics , Secretory Vesicles/genetics , Secretory Vesicles/immunology
5.
Front Cell Neurosci ; 14: 587072, 2020.
Article in English | MEDLINE | ID: mdl-33192329

ABSTRACT

Neurotransmitter release at retinal ribbon-style synapses utilizes a specialized t-SNARE protein called syntaxin3B (STX3B). In contrast to other syntaxins, STX3 proteins can be phosphorylated in vitro at T14 by Ca2+/calmodulin-dependent protein kinase II (CaMKII). This modification has the potential to modulate SNARE complex formation required for neurotransmitter release in an activity-dependent manner. To determine the extent to which T14 phosphorylation occurs in vivo in the mammalian retina and characterize the pathway responsible for the in vivo phosphorylation of T14, we utilized quantitative immunofluorescence to measure the levels of STX3 and STX3 phosphorylated at T14 (pSTX3) in the synaptic terminals of mouse retinal photoreceptors and rod bipolar cells (RBCs). Results demonstrate that STX3B phosphorylation at T14 is light-regulated and dependent upon the elevation of intraterminal Ca2+. In rod photoreceptor terminals, the ratio of pSTX3 to STX3 was significantly higher in dark-adapted mice, when rods are active, than in light-exposed mice. By contrast, in RBC terminals, the ratio of pSTX3 to STX3 was higher in light-exposed mice, when these terminals are active, than in dark-adapted mice. These results were recapitulated in the isolated eyecup preparation, but only when Ca2+ was included in the external medium. In the absence of external Ca2+, pSTX3 levels remained low regardless of light/dark exposure. Using the isolated RBC preparation, we next showed that elevation of intraterminal Ca2+ alone was sufficient to increase STX3 phosphorylation at T14. Furthermore, both the non-specific kinase inhibitor staurosporine and the selective CaMKII inhibitor AIP inhibited the Ca2+-dependent increase in the pSTX3/STX3 ratio in isolated RBC terminals, while in parallel experiments, AIP suppressed RBC depolarization-evoked exocytosis, measured using membrane capacitance measurements. Our data support a novel, illumination-regulated modulation of retinal ribbon-style synapse function in which activity-dependent Ca2+ entry drives the phosphorylation of STX3B at T14 by CaMKII, which in turn, modulates the ability to form SNARE complexes required for exocytosis.

6.
Int J Mol Sci ; 21(17)2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32882943

ABSTRACT

Calmodulin binding is a nearly universal property of gap junction proteins, imparting a calcium-dependent uncoupling behavior that can serve in an emergency to decouple a stressed cell from its neighbors. However, gap junctions that function as electrical synapses within networks of neurons routinely encounter large fluctuations in local cytoplasmic calcium concentration; frequent uncoupling would be impractical and counterproductive. We have studied the properties and functional consequences of calmodulin binding to the electrical synapse protein Connexin 35 (Cx35 or gjd2b), homologous to mammalian Connexin 36 (Cx36 or gjd2). We find that specializations in Cx35 calmodulin binding sites make it relatively impervious to moderately high levels of cytoplasmic calcium. Calmodulin binding to a site in the C-terminus causes uncoupling when calcium reaches low micromolar concentrations, a behavior prevented by mutations that eliminate calmodulin binding. However, milder stimuli promote calcium/calmodulin-dependent protein kinase II activity that potentiates coupling without interference from calmodulin binding. A second calmodulin binding site in the end of the Cx35 cytoplasmic loop, homologous to a calmodulin binding site present in many connexins, binds calmodulin with very low affinity and stoichiometry. Together, the calmodulin binding sites cause Cx35 to uncouple only at extreme levels of intracellular calcium.


Subject(s)
Calmodulin/metabolism , Connexins/metabolism , Electrical Synapses/physiology , Gap Junctions/physiology , Calcium Signaling , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calmodulin/genetics , Connexins/genetics , HeLa Cells , Humans , Mutation , Phosphorylation , Protein Binding , Protein Transport , Gap Junction delta-2 Protein
7.
Biophys J ; 118(4): 967-979, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31653448

ABSTRACT

First proposed as a specialized mode of release at sensory neurons possessing ribbon synapses, multivesicular release has since been described throughout the central nervous system. Many aspects of multivesicular release remain poorly understood. We explored mechanisms underlying simultaneous multivesicular release at ribbon synapses in salamander retinal rod photoreceptors. We assessed spontaneous release presynaptically by recording glutamate transporter anion currents (IA(glu)) in rods. Spontaneous IA(glu) events were correlated in amplitude and kinetics with simultaneously measured miniature excitatory postsynaptic currents in horizontal cells. Both measures indicated that a significant fraction of events is multiquantal, with an analysis of IA(glu) revealing that multivesicular release constitutes ∼30% of spontaneous release events. IA(glu) charge transfer increased linearly with event amplitude showing that larger events involve greater glutamate release. The kinetics of large and small IA(glu) events were identical as were rise times of large and small miniature excitatory postsynaptic currents, indicating that the release of multiple vesicles during large events is highly synchronized. Effects of exogenous Ca2+ buffers suggested that multiquantal, but not uniquantal, release occurs preferentially near Ca2+ channels clustered beneath synaptic ribbons. Photoinactivation of ribbons reduced the frequency of spontaneous multiquantal events without affecting uniquantal release frequency, showing that spontaneous multiquantal release requires functional ribbons. Although both occur at ribbon-style active zones, the absence of cross-depletion indicates that evoked and spontaneous multiquantal release from ribbons involve different vesicle pools. Introducing an inhibitory peptide into rods to interfere with the SNARE protein, syntaxin 3B, selectively reduced multiquantal event frequency. These results support the hypothesis that simultaneous multiquantal release from rods arises from homotypic fusion among neighboring vesicles on ribbons and involves syntaxin 3B.


Subject(s)
Synapses , Synaptic Vesicles , Excitatory Postsynaptic Potentials , Qa-SNARE Proteins , Retinal Rod Photoreceptor Cells , Synaptic Transmission
8.
J Biol Chem ; 294(9): 3012-3023, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30563839

ABSTRACT

Mast cells (MCs) participate in allergy, inflammation, and defense against pathogens. They release multiple immune mediators via exocytosis, a process that requires SNARE proteins, including syntaxins (Stxs). The identity of the Stxs involved in MC exocytosis remains controversial. Here, we studied the roles of Stx3 and -4 in fully developed MCs from conditional knockout mice by electrophysiology and EM, and found that Stx3, and not Stx4, is crucial for MC exocytosis. The main defect seen in Stx3-deficient MCs was their inability to engage multigranular compound exocytosis, while leaving most single-vesicle fusion events intact. We used this defect to show that this form of exocytosis is not only required to accelerate MC degranulation but also essential to achieve full degranulation. The exocytic defect was severe but not absolute, indicating that an Stx other than Stx3 and -4 is also required for exocytosis in MCs. The removal of Stx3 affected only regulated exocytosis, leaving other MC effector responses intact, including the secretion of cytokines via constitutive exocytosis. Our in vivo model of passive systemic anaphylaxis showed that the residual exocytic function of Stx3-deficient MCs was sufficient to drive a full anaphylactic response in mice.


Subject(s)
Exocytosis , Mast Cells/cytology , Qa-SNARE Proteins/metabolism , Animals , Cell Count , Cell Degranulation , Cell Differentiation , Gene Knockout Techniques , Kinetics , Mice , Qa-SNARE Proteins/deficiency , Qa-SNARE Proteins/genetics
9.
J Biol Chem ; 293(19): 7148-7159, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29599294

ABSTRACT

Mast cells (MCs) play pivotal roles in many inflammatory conditions including infections, anaphylaxis, and asthma. MCs store immunoregulatory compounds in their large cytoplasmic granules and, upon stimulation, secrete them via regulated exocytosis. Exocytosis in many cells requires the participation of Munc18 proteins (also known as syntaxin-binding proteins), and we found that mature MCs express all three mammalian isoforms: Munc18-1, -2, and -3. To study their functions in MC effector responses and test the role of MC degranulation in anaphylaxis, we used conditional knockout (cKO) mice in which each Munc18 protein was deleted exclusively in MCs. Using recordings of plasma membrane capacitance for high-resolution analysis of exocytosis in individual MCs, we observed an almost complete absence of exocytosis in Munc18-2-deficient MCs but intact exocytosis in MCs lacking Munc18-1 or Munc18-3. Stereological analysis of EM images of stimulated MCs revealed that the deletion of Munc18-2 also abolishes the homotypic membrane fusion required for compound exocytosis. We confirmed the severe defect in regulated exocytosis in the absence of Munc18-2 by measuring the secretion of mediators stored in MC granules. Munc18-2 cKO mice had normal morphology, development, and distribution of their MCs, indicating that Munc18-2 is not essential for the migration, retention, and maturation of MC-committed progenitors. Despite that, we found that Munc18-2 cKO mice were significantly protected from anaphylaxis. In conclusion, MC-regulated exocytosis is required for the anaphylactic response, and Munc18-2 is the sole Munc18 isoform that mediates membrane fusion during MC degranulation.


Subject(s)
Exocytosis/physiology , Mast Cells/metabolism , Munc18 Proteins/physiology , Anaphylaxis/physiopathology , Animals , Cell Degranulation , Gene Deletion , Mast Cells/ultrastructure , Membrane Fusion/physiology , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Munc18 Proteins/genetics , Patch-Clamp Techniques
10.
J Biol Chem ; 293(1): 345-358, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29141910

ABSTRACT

Mast cells (MCs) are involved in host defenses against pathogens and inflammation. Stimulated MCs release substances stored in their granules via regulated exocytosis. In other cell types, Munc13 (mammalian homolog of Caenorhabditis elegans uncoordinated gene 13) proteins play essential roles in regulated exocytosis. Here, we found that MCs express Munc13-2 and -4, and we studied their roles using global and conditional knock-out (KO) mice. In a model of systemic anaphylaxis, we found no difference between WT and Munc13-2 KO mice, but global and MC-specific Munc13-4 KO mice developed less hypothermia. This protection correlated with lower plasma histamine levels and with histological evidence of defective MC degranulation but not with changes in MC development, distribution, numbers, or morphology. In vitro assays revealed that the defective response in Munc13-4-deficient MCs was limited to regulated exocytosis, leaving other MC secretory effector responses intact. Single cell capacitance measurements in MCs from mouse mutants differing in Munc13-4 expression levels in their MCs revealed that as levels of Munc13-4 decrease, the rate of exocytosis declines first, and then the total amount of exocytosis decreases. A requirement for Munc13-2 in MC exocytosis was revealed only in the absence of Munc13-4. Electrophysiology and EM studies uncovered that the number of multigranular compound events (i.e. granule-to-granule homotypic fusion) was severely reduced in the absence of Munc13-4. We conclude that although Munc13-2 plays a minor role, Munc13-4 is essential for regulated exocytosis in MCs, and that this MC effector response is required for a full anaphylactic response.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Anaphylaxis , Animals , Disease Models, Animal , Exocytosis/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Mast Cells/metabolism , Mast Cells/physiology , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Protein Isoforms , Protein Transport
11.
Biophys J ; 113(10): 2281-2298, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28863864

ABSTRACT

Neurons that form ribbon-style synapses are specialized for continuous exocytosis. To this end, their synaptic terminals contain numerous synaptic vesicles, some of which are ribbon associated, that have difference susceptibilities for undergoing Ca2+-dependent exocytosis. In this study, we probed the relationship between previously defined vesicle populations and determined their fusion competency with respect to SNARE complex formation. We found that both the rapidly releasing vesicle pool and the releasable vesicle pool of the retinal bipolar cell are situated at the ribbon-style active zones, where they functionally interact. A peptide inhibitor of SNARE complex formation failed to block exocytosis from either pool, suggesting that these two vesicle pools have formed the SNARE complexes necessary for fusion. By contrast, a third, slower component of exocytosis was blocked by the peptide, as was the functional replenishment of vesicle pools, indicating that few vesicles outside of the ribbon-style active zones were initially fusion competent. In cone photoreceptors, similar to bipolar cells, fusion of the initial ribbon-associated synaptic vesicle cohort was not blocked by the SNARE complex-inhibiting peptide, whereas a later phase of exocytosis, attributable to the recruitment and subsequent fusion of vesicles newly arrived at the synaptic ribbons, was blocked. Together, our results support a model in which stimulus-evoked exocytosis in retinal ribbon synapses is SNARE-dependent; where vesicles higher up on the synaptic ribbon replenish the rapidly releasing vesicle pool; and at any given time, there are sufficient SNARE complexes to support the fusion of the entire ribbon-associated cohort of vesicles. An important implication of these results is that ribbon-associated vesicles can form intervesicular SNARE complexes, providing mechanistic insight into compound fusion at ribbon-style synapses.


Subject(s)
Synaptic Vesicles/metabolism , Animals , Calcium/metabolism , Exocytosis , Goldfish , Neurons/cytology , Retina/cytology , SNARE Proteins/metabolism
12.
Mol Cell Neurosci ; 60: 53-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24680688

ABSTRACT

Ribbon synapses in the retina lack the t-SNARE (target-soluble N-ethylmaleimide-sensitive factor attachment protein receptor) syntaxin 1A that is found in conventional synapses of the nervous system, but instead contain the related isoform syntaxin 3B. Previous studies have demonstrated that syntaxin 3B is essential for synaptic vesicle exocytosis in ribbon synapses, but syntaxin 3B is less efficient than syntaxin 1A in binding the t-SNARE protein SNAP-25 and catalyzing vesicle fusion. We demonstrate here that syntaxin 3B is localized mainly on the presynaptic membrane of retinal ribbon synapses and that a subset of syntaxin 3B is localized in close proximity to the synaptic ribbon. We show further, that syntaxin 3B can be phosphorylated by the Ca(2+)/calmodulin-dependent protein kinase II (CaMKII). We determine that the phosphorylation site is located close to the N-terminus at T14. Syntaxin 3B with a phosphomimetic mutation (T14E) had a stronger binding affinity for SNAP-25 compared with wild type syntaxin 3B. We propose that phosphorylation of syntaxin 3B by CaMKII can modulate the assembly of the SNARE complex in ribbon synapses of the retina, and might regulate the exocytosis of synaptic vesicles in ribbon synapses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Protein Processing, Post-Translational , Qa-SNARE Proteins/metabolism , Synaptosomal-Associated Protein 25/metabolism , Animals , Binding Sites , Exocytosis , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Presynaptic Terminals/metabolism , Protein Binding , Protein Transport , Qa-SNARE Proteins/chemistry , Qa-SNARE Proteins/genetics , Retina/metabolism
13.
J Neurophysiol ; 108(11): 3059-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22972962

ABSTRACT

Ca(2+) signaling in synaptic terminals plays a critical role in neurotransmitter release and short-term synaptic plasticity. In the present study, we examined the role of synaptic Ca(2+) handling mechanisms in the synaptic terminals of mammalian rod bipolar cells, neurons that play a pivotal role in the high-sensitivity vision pathway. We found that mitochondria sequester Ca(2+) under conditions of high Ca(2+) load, maintaining intraterminal Ca(2+) near resting levels. Indeed, the effect of the mitochondria was so powerful that the ability to clamp intraterminal Ca(2+) with a somatically positioned whole cell patch pipette was compromised. The plasma membrane Ca(2+)-ATPase (PMCA), but not the Na(+)/Ca(2+) exchanger (NCX) or the sarco(endo)plasmic reticulum Ca(2+)-ATPase (SERCA), was an important regulator of resting Ca(2+). Furthermore, PMCA activity, but not NCX or SERCA activity, was essential for the recovery of Ca(2+) levels following depolarization-evoked Ca(2+) entry. Loss of PMCA function was also associated with impaired restoration of membrane surface area following depolarization-evoked exocytosis. Given its roles in the regulation of intraterminal Ca(2+) at rest and after a stimulus-evoked Ca(2+) rise, the PMCA is poised to modulate luminance coding and adaptation to background illumination in the mammalian rod bipolar cell.


Subject(s)
Calcium Signaling , Calcium/metabolism , Presynaptic Terminals/metabolism , Action Potentials , Animals , Exocytosis , Goldfish , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Plasma Membrane Calcium-Transporting ATPases/metabolism , Retinal Rod Photoreceptor Cells/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sodium-Calcium Exchanger/metabolism
14.
Invest Ophthalmol Vis Sci ; 52(6): 3083-8, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21310914

ABSTRACT

PURPOSE: Previously, retinopetal axons containing histamine and dopaminergic neurons expressing histamine H(1)-receptor had been localized in mouse retinas using anatomic techniques. The goal of these experiments was to demonstrate that these receptors are functional. METHODS: Dopaminergic cells were acutely isolated from retinas of transgenic mice expressing red fluorescent protein under control of the tyrosine hydroxylase promoter and loaded with the calcium indicator Fura-2. RESULTS: Under control conditions, there were spontaneous oscillations in the levels of free intracellular calcium in dopaminergic cells. These oscillations were abolished in nominally calcium-free extracellular medium and in 1 µM tetrodotoxin, findings suggesting that the oscillations were mediated by calcium entry across the plasma membrane in response to sodium-dependent action potentials. Histamine increased the mean free intracellular calcium in the dopaminergic cells by increasing the frequency and/or amplitude of the calcium oscillations. The effects of histamine were dose-dependent and reached maximum at 5 µM. With this dose, there was a 65% increase in the mean free intracellular calcium concentration. The histamine H(1)-receptor antagonist, pyrilamine, blocked the effects of 5 µM histamine when applied at 50 µM. The selective histamine H(1)-receptor agonists, 2-(3-trifluoromethylphenyl) histamine and methylhistaprodifen significantly increased mean free intracellular calcium when applied at 5 µM. CONCLUSIONS: Histamine released from retinopetal axons in the mouse retina can elevate intracellular calcium levels in the perikarya of dopaminergic cells via the activation of histamine H(1)-receptors.


Subject(s)
Axons/drug effects , Calcium/metabolism , Dopamine/metabolism , Histamine/pharmacology , Receptors, Histamine H1/metabolism , Retina/drug effects , Animals , Axons/metabolism , Dose-Response Relationship, Drug , Histamine Agonists/pharmacology , Histamine H1 Antagonists/pharmacology , Mice , Mice, Transgenic , Retina/metabolism
15.
Vis Neurosci ; 28(1): 109-19, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21272392

ABSTRACT

Bipolar cells play a vital role in the transfer of visual information across the vertebrate retina. The synaptic output of these neurons is regulated by factors that are extrinsic and intrinsic. Relatively little is known about the intrinsic factors that regulate neurotransmitter exocytosis. Much of what we know about intrinsic presynaptic mechanisms that regulate glutamate release has come from the study of the unusually large and accessible synaptic terminal of the goldfish rod-dominant bipolar cell, the Mb1 bipolar cell. However, over the past several years, examination of presynaptic mechanisms governing neurotransmitter release has been extended to the mammalian rod bipolar cell. In this review, we discuss the recent advances in our understanding of synaptic vesicle dynamics and neurotransmitter release in rodent rod bipolar cells and consider how these properties help to shape the synaptic output of the mammalian retina.


Subject(s)
Presynaptic Terminals/physiology , Retinal Bipolar Cells/physiology , Synapses/physiology , Animals , Calcium Signaling/physiology , Cytoplasm/physiology , Endocytosis/physiology , Excitatory Postsynaptic Potentials/physiology , Humans , Retina/cytology , Retina/physiology , Synaptic Vesicles/physiology
16.
Neuron ; 66(6): 884-95, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20620874

ABSTRACT

Synaptic vesicle 2 (SV2) proteins, critical for proper nervous system function, are implicated in human epilepsy, yet little is known about their function. We demonstrate, using direct approaches, that loss of the major SV2 isoform in a central nervous system nerve terminal is associated with an elevation in both resting and evoked presynaptic Ca(2+) signals. This increase is essential for the expression of the SV2B(-/-) secretory phenotype, characterized by changes in synaptic vesicle dynamics, synaptic plasticity, and synaptic strength. Short-term reproduction of the Ca(2+) phenotype in wild-type nerve terminals reproduces almost all aspects of the SV2B(-/-) secretory phenotype, while rescue of the Ca(2+) phenotype in SV2B(-/-) neurons relieves every facet of the SV2B(-/-) secretory phenotype. Thus, SV2 controls key aspects of synaptic functionality via its ability to regulate presynaptic Ca(2+), suggesting a potential new target for therapeutic intervention in the treatment of epilepsy.


Subject(s)
Calcium/metabolism , Membrane Glycoproteins/physiology , Nerve Tissue Proteins/physiology , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Alcohol Oxidoreductases , Analysis of Variance , Animals , Biophysics , Calcium Signaling/genetics , Calcium Signaling/physiology , Chelating Agents/pharmacology , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Egtazic Acid/pharmacology , Electric Stimulation/methods , Membrane Glycoproteins/deficiency , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission/methods , Nerve Tissue Proteins/deficiency , Patch-Clamp Techniques/methods , Phosphoproteins/metabolism , Presynaptic Terminals/ultrastructure , Protein Kinase C-alpha/metabolism , Retina/cytology , Retinal Rod Photoreceptor Cells/ultrastructure
17.
Biophys J ; 98(10): 2102-10, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20483317

ABSTRACT

Exocytosis from the rod photoreceptor is stimulated by submicromolar Ca(2+) and exhibits an unusually shallow dependence on presynaptic Ca(2+). To provide a quantitative description of the photoreceptor Ca(2+) sensor for exocytosis, we tested a family of conventional and allosteric computational models describing the final Ca(2+)-binding steps leading to exocytosis. Simulations were fit to two measures of release, evoked by flash-photolysis of caged Ca(2+): exocytotic capacitance changes from individual rods and postsynaptic currents of second-order neurons. The best simulations supported the occupancy of only two Ca(2+) binding sites on the rod Ca(2+) sensor rather than the typical four or five. For most models, the on-rates for Ca(2+) binding and maximal fusion rate were comparable to those of other neurons. However, the off-rates for Ca(2+) unbinding were unexpectedly slow. In addition to contributing to the high-affinity of the photoreceptor Ca(2+) sensor, slow Ca(2+) unbinding may support the fusion of vesicles located at a distance from Ca(2+) channels. In addition, partial sensor occupancy due to slow unbinding may contribute to the linearization of the first synapse in vision.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Photoreceptor Cells/physiology , Retinal Cone Photoreceptor Cells/physiology , Synapses/physiology , Synaptic Transmission/physiology , Vision, Ocular/physiology , Action Potentials/physiology , Animals , Calcium Signaling/physiology , Electric Capacitance , Electric Stimulation , Electrophysiology/methods , Exocytosis/physiology , Membrane Potentials/physiology , Neural Inhibition , Neural Pathways , Neurons/physiology , Patch-Clamp Techniques , Retinal Horizontal Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Urodela
18.
J Biol Chem ; 284(29): 19445-51, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19473977

ABSTRACT

Mast cell degranulation is a highly regulated, calcium-dependent process, which is important for the acute release of inflammatory mediators during the course of many pathological conditions. We previously found that Synaptotagmin-2, a calcium sensor in neuronal exocytosis, was expressed in a mast cell line. We postulated that this protein may be involved in the control of mast cell-regulated exocytosis, and we generated Synaptotagmin-2 knock-out mice to test our hypothesis. Mast cells from this mutant animal conferred an abnormally decreased passive cutaneous anaphylaxis reaction on mast cell-deficient mice that correlated with a specific defect in mast cell-regulated exocytosis, leaving constitutive exocytosis and nonexocytic mast cell effector responses intact. This defect was not secondary to abnormalities in the development, maturation, migration, morphology, synthesis, and storage of inflammatory mediators, or intracellular calcium transients of the mast cells. Unlike neurons, the lack of Synaptotagmin-2 in mast cells was not associated with increased spontaneous exocytosis.


Subject(s)
Exocytosis , Mast Cells/metabolism , Synaptotagmin II/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Calcium/metabolism , Cell Count , Cell Differentiation , Cells, Cultured , Cytoplasmic Granules/metabolism , Female , Hypersensitivity/genetics , Hypersensitivity/metabolism , Immunoblotting , Immunohistochemistry , Male , Mast Cells/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Synaptotagmin II/genetics
19.
Vis Neurosci ; 25(4): 523-33, 2008.
Article in English | MEDLINE | ID: mdl-18764958

ABSTRACT

To better understand synaptic signaling at the mammalian rod bipolar cell terminal and pave the way for applying genetic approaches to the study of visual information processing in the mammalian retina, synaptic vesicle dynamics and intraterminal calcium were monitored in terminals of acutely isolated mouse rod bipolar cells and the number of ribbon-style active zones quantified. We identified a releasable pool, corresponding to a maximum of 7 s. The presence of a smaller, rapidly releasing pool and a small, fast component of refilling was also suggested. Following calcium channel closure, membrane surface area was restored to baseline with a time constant that ranged from 2 to 21 s depending on the magnitude of the preceding Ca2+ transient. In addition, a brief, calcium-dependent delay often preceded the start of onset of membrane recovery. Thus, several aspects of synaptic vesicle dynamics appear to be conserved between rod-dominant bipolar cells of fish and mammalian rod bipolar cells. A major difference is that the number of vesicles available for release is significantly smaller in the mouse rod bipolar cell, both as a function of the total number per neuron and on a per active zone basis.


Subject(s)
Mice , Retinal Bipolar Cells/physiology , Retinal Rod Photoreceptor Cells/physiology , Synaptic Vesicles/physiology , Animals , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling , Electrophysiological Phenomena , Endocytosis , Exocytosis , Immunohistochemistry , Mice, Inbred C57BL , Microscopy, Confocal , Retinal Bipolar Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Synaptic Vesicles/metabolism , Time Factors
20.
J Neurophysiol ; 99(1): 25-36, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17989244

ABSTRACT

Time-resolved capacitance measurements in combination with fluorescence measurements of internal calcium suggested three kinetic components of release in acutely isolated cone photoreceptors of the tiger salamander. A 45-fF releasable pool, corresponding to about 1,000 vesicles, was identified. This pool could be depleted with a time constant of a few hundred milliseconds and its recovery from depletion was quite rapid (tau approximately 1 s). The fusion of vesicles in this pool was blocked by low-millimolar EGTA. Endocytosis was sufficiently slow that it is likely that refilling of the releasable pool occurred from preformed vesicles. A second, slower component of release (tau(depletion) approximately 3 s) was identified that was approximately twice the size of the releasable pool. This pool may serve as a first reserve pool that replenishes the releasable pool. Computer simulations indicate that the properties of the releasable and first reserve pools are sufficient to maintain synaptic signaling for several seconds in the face of near-maximal stimulations and in the absence of other sources of vesicles. Along with lower rates of depletion, additional mechanisms, such as replenishment from distal reserve pools and the fast recycling of vesicles, may further contribute to the maintenance of graded, tonic release from cone photoreceptors.


Subject(s)
Glutamic Acid/metabolism , Retina/physiology , Retinal Cone Photoreceptor Cells/physiology , Synapses/metabolism , Synaptic Transmission/physiology , Synaptic Vesicles/metabolism , Ambystoma , Animals , Calcium Signaling/physiology , Computer Simulation , Electric Stimulation , Endocytosis/physiology , Exocytosis/physiology , Membrane Fusion/physiology , Patch-Clamp Techniques , Reaction Time/physiology , Retina/ultrastructure , Retinal Cone Photoreceptor Cells/ultrastructure , Time Factors , Vision, Ocular/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...