Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 388
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(29): e2409605121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985768

ABSTRACT

Members of the synaptophysin and synaptogyrin family are vesicle proteins with four transmembrane domains. In spite of their abundance in synaptic vesicle (SV) membranes, their role remains elusive and only mild defects at the cellular and organismal level are observed in mice lacking one or more family members. Here, we show that coexpression with synapsin in fibroblasts of each of the four brain-enriched members of this family-synaptophysin, synaptoporin, synaptogyrin 1, and synaptogyrin 3-is sufficient to generate clusters of small vesicles in the same size range of SVs. Moreover, mice lacking all these four proteins have larger SVs. We conclude that synaptophysin and synaptogyrin family proteins play an overlapping function in the biogenesis of SVs and in determining their small size.


Subject(s)
Synaptic Vesicles , Synaptogyrins , Synaptophysin , Animals , Synaptophysin/metabolism , Synaptophysin/genetics , Synaptic Vesicles/metabolism , Mice , Synaptogyrins/metabolism , Synaptogyrins/genetics , Synapsins/metabolism , Synapsins/genetics , Mice, Knockout , Fibroblasts/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Rats , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics
2.
Proc Natl Acad Sci U S A ; 120(45): e2311484120, 2023 Nov 07.
Article in English | MEDLINE | ID: mdl-37903271

ABSTRACT

The synaptic vesicle protein Synaptophysin (Syp) has long been known to form a complex with the Vesicle associated soluble N-ethylmaleimide sensitive fusion protein attachment receptor (v-SNARE) Vesicle associated membrane protein (VAMP), but a more specific molecular function or mechanism of action in exocytosis has been lacking because gene knockouts have minimal effects. Utilizing fully defined reconstitution and single-molecule measurements, we now report that Syp functions as a chaperone that determines the number of SNAREpins assembling between a ready-release vesicle and its target membrane bilayer. Specifically, Syp directs the assembly of 12 ± 1 SNAREpins under each docked vesicle, even in the face of an excess of SNARE proteins. The SNAREpins assemble in successive waves of 6 ± 1 and 5 ± 2 SNAREpins, respectively, tightly linked to oligomerization of and binding to the vesicle Ca++ sensor Synaptotagmin. Templating of 12 SNAREpins by Syp is likely the direct result of its hexamer structure and its binding of VAMP2 dimers, both of which we demonstrate in detergent extracts and lipid bilayers.


Subject(s)
Membrane Fusion , Synaptic Vesicles , Synaptophysin/genetics , Synaptophysin/metabolism , Membrane Fusion/physiology , Synaptic Vesicles/metabolism , Synaptotagmins/metabolism , SNARE Proteins/metabolism , Exocytosis/physiology
3.
J Cell Biochem ; 125(3): e30529, 2024 03.
Article in English | MEDLINE | ID: mdl-38308620

ABSTRACT

Sept8 is a vesicle associated protein and there are two typical transcriptional variants (Sept8-204 and Sept8-201) expressed in mice brain. Interestingly, the coexpression of Sept8-204/Sept5 induces the formation of small sized vesicle-like structure, while that of the Sept8-201/Sept5 produces large puncta. Sept8 is previously shown to be palmitoylated. Here it was further revealed that protein palmitoylation is required for Sept8-204/Sept5 to maintain small sized vesicle-like structure and colocalize with synaptophysin, since either the expression of nonpalmitoylated Sept8-204 mutant (Sept8-204-3CA) or inhibiting Sept8-204 palmitoylation by 2-BP with Sept5 produces large puncta, which barely colocalizes with synaptophysin (SYP). Moreover, it was shown that the dynamic palmitoylation of Sept8-204 is controlled by ZDHHC17 and PPT1, loss of ZDHHC17 decreases Sept8-204 palmitoylation and induces large puncta, while loss of PPT1 increases Sept8-204 palmitoylation and induces small sized vesicle-like structure. Together, these findings suggest that palmitoylation is essential for the maintenance of the small sized vesicle-like structure for Sept8-204/Sept5, and may hint their important roles in synaptic functions.


Subject(s)
Lipoylation , Septins , Animals , Mice , Cell Cycle Proteins/metabolism , Septins/genetics , Septins/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
4.
IUBMB Life ; 76(8): 548-562, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38390757

ABSTRACT

Age-related reduction in spine density, synaptic marker expression, and synaptic efficiency are frequently reported. These changes provide the cellular and molecular basis for the cognitive decline characteristic for old age. Nevertheless, there are several approaches that have the potential to ameliorate these processes and improve cognition, caloric restriction being one of the most promising and widely studied. While lifelong caloric restriction is known for its numerous beneficial effects, including improved cognitive abilities and increased expression of proteins essential for synaptic structure and function, the effects of late-onset and/or short-term CR on synaptic plasticity have yet to be investigated. We have previously documented that the effects of CR are strongly dependent on whether CR is initiated in young or old subjects. With this in mind, we conducted a long-term study in aging Wistar rats to examine changes in the expression of several key synaptic markers under the regimen of CR started at different time points in life. We found a significant increase in the expression of both presynaptic and postsynaptic markers. However, taking into account previously reported changes in the behavior detected in these animals, we consider that this increase cannot represent beneficial effect of CR.


Subject(s)
Caloric Restriction , Neuronal Plasticity , Animals , Male , Rats , Age Factors , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Cadherins/genetics , Cadherins/metabolism , Diet , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , GAP-43 Protein/genetics , GAP-43 Protein/metabolism , Gene Expression Regulation/physiology , Neuronal Plasticity/physiology , Rats, Wistar , RNA, Messenger/genetics , RNA, Messenger/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
5.
Hum Mol Genet ; 30(8): 629-643, 2021 05 17.
Article in English | MEDLINE | ID: mdl-33693569

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive degenerative motor neuron disease characterized by symmetrical muscle weakness and atrophy of limb and trunk muscles being the most severe genetic disease in children. In SMA mouse models, motor nerve terminals display neurotransmitter release reduction, endocytosis decrease and mitochondria alterations. The relationship between these changes is, however, not well understood. In the present study, we investigated whether the endocytosis impairment could be related to the functional alteration of the presynaptic mitochondria during action potential (AP) firing. To this aim, we generated a Synaptophysin-pHluorin (SypHy) transgenic mouse, crossed it with Taiwanese SMA mice, and recorded exo- and endocytosis and mitochondria Ca2+ signaling in real-time at ex vivo motor nerve terminals of Taiwanese-SypHy mice. The experiments were performed at the beginning of the motor symptoms to get an integrated view of the nerve terminal's functional state before degeneration. Our electrophysiological and live imaging results demonstrated that the mitochondria's capacity to increase matrix-free Ca2+ in SMA mice was significantly limited during nerve AP firing, except when the rate of Ca2+ entry to the cytosol was considerably reduced. These results indicate that both the mitochondrial Ca2+ signaling alterations and the secretion machinery defects are significant players in the dysfunction of the presynaptic terminal in SMA.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Motor Neurons/physiology , Muscular Atrophy, Spinal/metabolism , Presynaptic Terminals/metabolism , Synaptic Transmission/physiology , Action Potentials/genetics , Action Potentials/physiology , Animals , Disease Models, Animal , Endocytosis/genetics , Endocytosis/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice, Transgenic , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/physiopathology , Synapses/genetics , Synapses/metabolism , Synapses/physiology , Synaptophysin/genetics , Synaptophysin/metabolism
6.
Differentiation ; 128: 43-56, 2022.
Article in English | MEDLINE | ID: mdl-36306693

ABSTRACT

Major histocompatibility complex class I (MHC-I) has been implicated in several types of neuroplasticity phenomena. Interferon beta-1b (IFN-ß) increases MHC-I expression by motoneurons after sciatic nerve crush in mice, improving axonal growth and functional recovery. Additionally, IFN-ß induces glial hypertrophy associated with upregulation of glial fibrillary acidic protein (GFAP) and MHC-I in murine astrocytes in vitro. As knowledge about MHC-I and its role in synaptic plasticity in human astrocytes (HAs) is scarce, we investigated these aspects in mature HAs obtained from the neocortex of patients undergoing surgery due to hippocampal sclerosis. Cells were exposed to media in the absence (0 IU/ml) or presence of IFN-ß for 5 days (500 IU/ml). Beta-2 microglobulin (ß2m), a component of the MHC-I, GFAP and vimentin proteins, was quantified by flow cytometry (FC) and increased by 100%, 60% and 46%, respectively, after IFN-ß exposure. We also performed qRT-PCR gene expression analyses for ß2m, GFAP, vimentin, and pro- and anti-inflammatory cytokines. Our data showed that IFN-ß-treated astrocytes displayed ß2m and GFAP gene upregulation. Additionally, they presented a proinflammatory profile with increase in the IL-6 and IL-1ß genes and a tendency to upregulate TNF-α. Moreover, we evaluated the effect of HAs conditioned medium (CM) on the formation/maintenance of neurites/synapses by the PC12 lineage. Synaptophysin protein expression was quantified by FC. The CM of IFN-ß-activated astrocytes was not harmful to PC12 neurites, and there was no change in synaptophysin protein expression. Therefore, IFN-ß activated HAs by increasing GFAP, vimentin and MHC-I protein expression. Like MHC-I modulation and astrocyte activation may be protective after peripheral nerve damage and in some neurodegenerative conditions, this study opens perspectives on the pathophysiological roles of astroglial MHC-I in the human CNS.


Subject(s)
Astrocytes , Interferon-beta , Humans , Animals , Mice , Astrocytes/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Synaptophysin/pharmacology , Vimentin/genetics , Vimentin/metabolism , Vimentin/pharmacology , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-beta/pharmacology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Major Histocompatibility Complex , Phenotype
7.
Morphologie ; 107(356): 67-79, 2023 Mar.
Article in English | MEDLINE | ID: mdl-35715368

ABSTRACT

Adverse experiences during pregnancy have a negative impact on the neuronal structure and behavior of offspring, but the effects of a father's life events on the outcome of progeny are scarce. The present study is intended to investigate whether paternal stress affects the offspring brain structure, especially those regions concerned with learning and formation of memory, namely the hippocampus (HC) and prefrontal cortex (PFC), and also the expression of certain genes linked to learning and memory in the offspring. Induced stress to male rats by five stressors, one per day followed by allowing them to mate with the normal, unstressed female. Synaptophysin immunoreactivity was assessed in the tissue sections of the HC and PFC as well as expression of genes concerned with learning and memory was evaluated by RT-PCR in the progeny of stress-received males. The progeny of stressed rats had reduced antisynaptophysin immunoreactivity in the HC and PFC. The synaptic density in HC was less in the A-S (Offspring of male rats who received stress during adulthood) and PA-S (offspring of male rats who received stress during both adolescence and adulthood) than in P-S (offspring of male rats who received stress during adolescence) and C-C (offspring of control) groups. Similar results were observed even in the PFC. The results of post hoc tests proved that the HC and PFC of the progeny of stress-exposed rats exhibited considerably less synaptic density than control (P<0.05), and the levels of expression of GAP-43, GRIN1, M1, and SYP genes in HC and PFC were down-regulated. This study concludes that paternal adverse experiences can affect the offspring's synaptic plasticity and also the genes, which can regulate learning and formation of memory.


Subject(s)
Hippocampus , Prefrontal Cortex , Pregnancy , Rats , Animals , Male , Female , Humans , GAP-43 Protein/metabolism , GAP-43 Protein/pharmacology , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Learning , Fathers , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Synaptophysin/pharmacology
8.
J Neurosci ; 41(16): 3563-3578, 2021 04 21.
Article in English | MEDLINE | ID: mdl-33664131

ABSTRACT

Synaptophysin (syp) is a major integral membrane protein of secretory vesicles. Previous work has demonstrated functions for syp in synaptic vesicle cycling, endocytosis, and synaptic plasticity, but the role of syp in the process of membrane fusion during Ca2+-triggered exocytosis remains poorly understood. Furthermore, although syp resides on both large dense-core and small synaptic vesicles, its role in dense-core vesicle function has received less attention compared with synaptic vesicle function. To explore the role of syp in membrane fusion and dense-core vesicle function, we used amperometry to measure catecholamine release from single vesicles in male and female mouse chromaffin cells with altered levels of syp and the related tetraspanner protein synaptogyrin (syg). Knocking out syp slightly reduced the frequency of vesicle fusion events below wild-type (WT) levels, but knocking out both syp and syg reduced the frequency 2-fold. Knocking out both proteins stabilized initial fusion pores, promoted fusion pore closure (kiss-and-run), and reduced late-stage fusion pore expansion. Introduction of a syp construct lacking its C-terminal dynamin-binding domain in syp knock-outs (KOs) increased the duration and fraction of kiss-and-run events, increased total catecholamine release per event, and reduced late-stage fusion pore expansion. These results demonstrated that syp and syg regulate dense-core vesicle function at multiple stages to initiate fusion, control the choice of mode between full-fusion and kiss-and-run, and influence the dynamics of both initial and late-stage fusion pores. The transmembrane domain (TMD) influences small initial fusion pores, and the C-terminal domain influences large late-stage fusion pores, possibly through an interaction with dynamin.SIGNIFICANCE STATEMENT The secretory vesicle protein synaptophysin (syp) is known to function in synaptic vesicle cycling, but its roles in dense-core vesicle functions, and in controlling membrane fusion during Ca2+-triggered exocytosis remain unclear. The present study used amperometry recording of catecholamine release from endocrine cells to assess the impact of syp and related proteins on membrane fusion. A detailed analysis of amperometric spikes arising from the exocytosis of single vesicles showed that these proteins influence fusion pores at multiple stages and control the choice between kiss-and-run and full-fusion. Experiments with a syp construct lacking its C terminus indicated that the transmembrane domain (TMD) influences the initial fusion pore, while the C-terminal domain influences later stages after fusion pore expansion.


Subject(s)
Chromaffin Cells/physiology , Exocytosis/physiology , Synaptophysin/physiology , Animals , Animals, Newborn , Catecholamines/metabolism , Dynamins/metabolism , Dynamins/physiology , Electrophysiological Phenomena , Exocytosis/genetics , Female , Membrane Fusion , Mice , Mice, Knockout , Pregnancy , Primary Cell Culture , Synaptic Vesicles/metabolism , Synaptic Vesicles/physiology , Synaptogyrins/genetics , Synaptogyrins/physiology , Synaptophysin/genetics
9.
J Biol Chem ; 296: 100266, 2021.
Article in English | MEDLINE | ID: mdl-33769286

ABSTRACT

The accurate retrieval of synaptic vesicle (SV) proteins during endocytosis is essential for the maintenance of neurotransmission. Synaptophysin (Syp) and synaptobrevin-II (SybII) are the most abundant proteins on SVs. Neurons lacking Syp display defects in the activity-dependent retrieval of SybII and a general slowing of SV endocytosis. To determine the role of the cytoplasmic C terminus of Syp in the control of these two events, we performed molecular replacement studies in primary cultures of Syp knockout neurons using genetically encoded reporters of SV cargo trafficking at physiological temperatures. Under these conditions, we discovered, 1) no slowing in SV endocytosis in Syp knockout neurons, and 2) a continued defect in SybII retrieval in knockout neurons expressing a form of Syp lacking its C terminus. Sequential truncations of the Syp C-terminus revealed a cryptic interaction site for the SNARE motif of SybII that was concealed in the full-length form. This suggests that a conformational change within the Syp C terminus is key to permitting SybII binding and thus its accurate retrieval. Furthermore, this study reveals that the sole presynaptic role of Syp is the control of SybII retrieval, since no defect in SV endocytosis kinetics was observed at physiological temperatures.


Subject(s)
Neurons/metabolism , Synaptic Vesicles/genetics , Synaptophysin/genetics , Vesicle-Associated Membrane Protein 2/genetics , Endocytosis/genetics , Gene Knockout Techniques , Hippocampus/metabolism , Hippocampus/pathology , Neurons/chemistry , Primary Cell Culture , SNARE Proteins/genetics , Synaptic Transmission/genetics , Synaptophysin/chemistry , Synaptosomes/chemistry , Synaptosomes/metabolism
10.
Cell Biol Toxicol ; 38(2): 347-369, 2022 04.
Article in English | MEDLINE | ID: mdl-33928466

ABSTRACT

Clinical surgical practices have found that children who undergo multiple anesthesia may have an increased risk of deficiencies in cognition and fine motor control. Here, we report that YT521-B homology domain family 1 (YTHDF1), a critical reader protein for N6-methyladenosine-modified mRNA, was significantly downregulated in the prefrontal cortex of young mice after multiple sevoflurane anesthesia exposures. Importantly, sevoflurane led to a decrease in protein synthesis in mouse cortical neurons that was fully rescued by YTHDF1, suggesting that anesthesia may affect early brain development by affecting m6A-dependent mRNA translation. Transcriptome-wide experiments showed that numerous mRNA targets related to synaptic functions in the prefrontal mouse cortex were associated with m6A methylation and YTHDF1. In particular, we found that synaptophysin, a critical presynaptic protein, was specifically modified by m6A methylation and associated with YTHDF1, and m6A methylation of synaptophysin decreased with multiple sevoflurane exposures. Importantly, we showed that fine motor control skills and cognitive functions were impaired in mice with multiple anesthesia exposures, and these effects were fully reversed by reintroducing YTHDF1 through a blood-brain barrier (BBB)-crossing viral delivery system. Finally, we found that the fine motor skills in children who underwent prolonged anesthesia were compromised 6 months after surgery. Our findings indicated that impairment in the translational regulation of mRNA via N6-methyladenosine methylation is a potential mechanism underlying the effects of anesthesia on neural development in the young brain. 1. N6-methyladenosine (m6A) modifications were involved in anesthesia-induced neurotoxicity. 2. Sevoflurane impairs m6A-mediated mRNA translation and leads to fine motor deficits in young mice. 3. YTHDF1, a m6A reader protein, rescued sevoflurane-induced protein synthesis inhibition and fine motor deficits in young mice.


Subject(s)
Adenosine , Protein Biosynthesis , Adenosine/genetics , Adenosine/metabolism , Animals , Cognition , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sevoflurane/adverse effects , Synaptophysin/genetics , Synaptophysin/metabolism
11.
Exp Cell Res ; 408(2): 112865, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34637763

ABSTRACT

Protein homeostasis (proteostasis) in multicellular organisms depends on the maintenance of force-bearing and force-generating cellular structures. Within myofibrillar Z-discs of striated muscle, isoforms of synaptopodin-2 (SYNPO2/myopodin) act as adapter proteins that are engaged in proteostasis of the actin-crosslinking protein filamin C (FLNc) under mechanical stress. SYNPO2 directly binds F-actin, FLNc and α-actinin and thus contributes to the architectural features of the actin cytoskeleton. By its association with autophagy mediating proteins, i.e. BAG3 and VPS18, SYNPO2 is also engaged in protein quality control and helps to target mechanical unfolded and damaged FLNc for degradation. Here we show that deficiency of all SYNPO2-isoforms in myotubes leads to decreased myofibrillar stability and deregulated autophagy under mechanical stress. In addition, isoform-specific proteostasis functions were revealed. The PDZ-domain containing variant SYNPO2b and the shorter, PDZ-less isoform SYNPO2e both localize to Z-discs. Yet, SYNPO2e is less stably associated with the Z-disc than SYNPO2b, and is dynamically transferred into FLNc-containing myofibrillar lesions under mechanical stress. SYNPO2e also recruits BAG3 into these lesions via interaction with the WW domain of BAG3. Our data provide evidence for a role of myofibrillar lesions as a transient quality control compartment essential to prevent and repair contraction-induced myofibril damage in muscle and indicate an important coordinating activity for SYNPO2 therein.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Microfilament Proteins/genetics , Muscle, Skeletal/metabolism , Stress, Mechanical , Vesicular Transport Proteins/genetics , Actin Cytoskeleton/genetics , Actinin/genetics , Actins/genetics , Animals , Autophagy/genetics , Cell Line , Cytoskeleton/genetics , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Striated/metabolism , Myofibrils/genetics , Myofibrils/metabolism , PDZ Domains/genetics , Protein Isoforms/genetics , Synaptophysin/genetics
12.
Alzheimers Dement ; 18(3): 523-528, 2022 03.
Article in English | MEDLINE | ID: mdl-34310040

ABSTRACT

INTRODUCTION: Synaptophysin, already related to X-linked intellectual disability, is expressed mainly in the central nervous system. Studies in humans indicate that the downregulation of synaptophysin could be involved in the development of dementia. Our study presents the first familial case of behavioral variant frontotemporal dementia associated with the co-occurrence of the repeat expansion in C9orf72 and a pathogenic variant in the SYP gene. METHODS: Exome sequencing and repeat-primed PCR for C9orf72 were performed for two siblings with clinical and imaging findings suggestive of slowly progressive behavioral frontotemporal dementia. RESULTS: We found that both siblings have the hexanucleotide expansion in C9orf72 and a null variant in the SYP gene. The most affected sibling presents the putative variant in a hemizygous state. With milder symptoms, his sister has the same pathogenic variant in heterozygosis, compatible with X-linked inheritance. DISCUSSION: Our results strengthened previous suggestive evidence that the phenotypes associated with C9orf72 repeat expansion are variable and probably influenced by additional genetic modifiers. We hypothesized that the pathogenic variant in the SYP gene might have modified the typical phenotype associated with the C9orf72 mutation.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/genetics , DNA Repeat Expansion/genetics , Frontotemporal Dementia/genetics , Humans , Mutation/genetics , Proteins/genetics , Synaptophysin/genetics
13.
Fish Physiol Biochem ; 48(2): 461-469, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35301620

ABSTRACT

Anadromous Pacific salmon (genus Oncorhynchus) are known for their homing behavior based on olfactory imprinting, which is formed during their seaward migration. Soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE/Snare) complex is a minimum unit of vesicle exocytosis from the pre-synaptic membrane. Its component genes (synaptosome-associated protein 25, syntaxin 1, and vesicle-associated membrane protein 2) are more strongly expressed in the olfactory nervous system (olfactory epithelium, olfactory bulb, and telencephalon) at the migration stages related to olfactory imprinting and/or retrieval in salmon. This study focused on the mRNA synthesis of synaptophysin (Syp), one of the Snare regulatory factors. syp is strongly expressed in chum salmon (Oncorhynchus keta) olfactory nervous system during the seaward migration and temporarily increased during the homeward migration. In reference to our previous studies, these expression changes were similar to the snare genes in the chum salmon. Therefore, syp and Snare component genes were synchronously expressed reflecting the development and short-term plasticity of the olfactory nervous system that is essential for olfactory imprinting.


Subject(s)
Oncorhynchus keta , Salmon , Animal Migration/physiology , Animals , Brain/metabolism , Exocytosis , Gene Expression , Oncorhynchus keta/genetics , Oncorhynchus keta/metabolism , SNARE Proteins/genetics , SNARE Proteins/metabolism , Salmon/genetics , Synaptophysin/genetics , Synaptophysin/metabolism
14.
J Neurochem ; 159(1): 78-89, 2021 10.
Article in English | MEDLINE | ID: mdl-34468992

ABSTRACT

Synaptobrevin-2 (Syb2) is a soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) that is essential for neurotransmitter release. It is the most numerous protein on a synaptic vesicle (SV) and drives SV fusion via interactions with its cognate SNARE partners on the presynaptic plasma membrane. Synaptophysin (Syp) is the second most abundant protein on SVs; however, in contrast to Syb2, it has no obligatory role in neurotransmission. Syp interacts with Syb2 on SVs, and the molecular nature of its interaction with Syb2 and its physiological role has been debated for decades. However, recent studies have revealed that the sole physiological role of Syp at the presynapse is to ensure the efficient retrieval of Syb2 during SV endocytosis. In this review, current theories surrounding the role of Syp in Syb2 trafficking will be discussed, in addition to the debate regarding the molecular nature of their interaction. A unifying model is presented that describes how Syp controls Syb2 function as part of an integrated mechanism involving key molecular players such as intersectin-1 and AP180/CALM. Finally, key future questions surrounding the role of Syp-dependent Syb2 trafficking will be posed, with respect to brain function in health and disease.


Subject(s)
Presynaptic Terminals/metabolism , Protein Transport/physiology , SNARE Proteins/metabolism , Synaptophysin/metabolism , Vesicle-Associated Membrane Protein 2/metabolism , Animals , Endocytosis/physiology , Humans , SNARE Proteins/genetics , Synapses/genetics , Synapses/metabolism , Synaptophysin/genetics , Vesicle-Associated Membrane Protein 2/genetics
15.
Exp Eye Res ; 213: 108807, 2021 12.
Article in English | MEDLINE | ID: mdl-34695438

ABSTRACT

The retinal circadian system consists of a network of clocks located virtually in every retinal cell-type. Although it is established that the circadian clock regulates many rhythmic processes in the retina, the links between retinal cell-specific clocks and visual function remain to be elucidated. Bmal1 is a principal, non-redundant component of the circadian clock in mammals and is required to keep 24 h rhythms in the retinal transcriptome and in visual processing under photopic light condition. In the current study, we investigated the retinal function in mice with a rod-specific knockout of Bmal1. For this purpose, we measured whole retina PER2::Luciferase bioluminescence and the dark-adapted electroretinogram (ERG). We observed circadian day-night differences in ERG a- and b-waves in control mice carrying one allele of Bmal1 in rods, with higher amplitudes during the subjective night. These differences were abolished in rod-specific Bmal1 knockout mice, whose ERG light-responses remained constitutively low (day-like). Overall, PER2::Luciferase rhythmicity in whole retinas was not defective in these mice but was characterized by longer period and higher rhythmic power compared to retinas with wild type Bmal1 gene. Taken together, these data suggest that a circadian clock located in rods regulates visual processing in a cell autonomous manner.


Subject(s)
Circadian Clocks/physiology , Dark Adaptation/physiology , Retinal Rod Photoreceptor Cells/metabolism , ARNTL Transcription Factors/genetics , Animals , Electroretinography , Female , Gene Expression Regulation/physiology , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Night Vision/physiology , Period Circadian Proteins/metabolism , Photic Stimulation , Real-Time Polymerase Chain Reaction , Retinal Rod Photoreceptor Cells/radiation effects , Rhodopsin/genetics , Synaptophysin/genetics
16.
Biochemistry (Mosc) ; 86(6): 704-715, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34225593

ABSTRACT

Early-life stress is a risk factor for the development of behavioral and cognitive disorders in humans and animals. Such stressful situations include social isolation in early postnatal ontogenesis. Behavioral and cognitive impairments associated with neuroplastic changes in brain structures. We have found that after ten weeks of social isolation, male Wistar rats show behavioral abnormalities and cognitive deficit, accompanied by an increase in the relative expression of gene encoding serine protease prolyl endopeptidase (PREP, EC 3.4.21.26) in the brain frontal cortex. The present study aimed to assess synaptophysin (SYP), brain-derived neurotrophic factor precursor (proBDNF), and PREP expression using Western blot in the brain structures - the hippocampus, frontal cortex, and striatum of the rats subjected to prolonged social isolation compared with group-housed animals. Twenty Wistar rats were used for this study (10 males and 10 females). Experimental animals (5 males and 5 females) were kept one per cage for nine months, starting from the age of one month. Ten-month-old socially isolated rats showed memory deficit in passive avoidance paradigm and Morris Water Maze and reactivity to novelty reduction. We used monoclonal antibodies for the Western blot analysis of the expression of SYP, proBDNF, and PREP in the rat brain structures. Social isolation caused a proBDNF expression reduction in the frontal cortex in females and a reduction in PREP expression in the striatum in males. These data suppose that neurotrophic factors and PREP are involved in the mechanisms of behavioral and cognitive impairments observed in the rats subjected to prolonged social isolation with an early life onset.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain/metabolism , Prolyl Oligopeptidases/genetics , Social Isolation , Stress, Psychological/metabolism , Animals , Female , Frontal Lobe/metabolism , Gene Expression Regulation , Hippocampus/metabolism , Male , Neuronal Plasticity , Rats , Rats, Wistar , Stress, Psychological/enzymology , Stress, Psychological/genetics , Synaptophysin/genetics
17.
J Neurovirol ; 26(4): 496-508, 2020 08.
Article in English | MEDLINE | ID: mdl-32394397

ABSTRACT

HIV-associated neurocognitive disorders (HAND) describe a spectrum of neuropsychological impairment caused by HIV-1 infection. While the sequence of cellular and physiological events that lead to HAND remains obscure, it likely involves chronic neuroinflammation. Host genetic markers that increase the risk for HAND have been reported, but replication of such studies is lacking, possibly due to inconsistent application of a behavioral phenotype across studies. In the current study, we used histopathologic phenotypes in order to validate putative risk alleles for HAND. The National NeuroAIDS Tissue Consortium, a longitudinal study of the neurologic manifestations of HIV. Data and specimens were obtained from 175 HIV-infected adults. After determining several potential covariates of neurocognitive functioning, we quantified levels of six histopathological markers in the frontal lobe in association with neurocognitive functioning: SYP, MAP 2, HLA-DR, Iba1, GFAP, and ß-amyloid. We then determined alleles of 15 candidate genes for their associations with neurocognitive functioning and histopathological markers. Finally, we identified the most plausible causal pathway based on our data using a multi-stage linear regression-based mediation analysis approach. None of the genetic markers were associated with neurocognitive functioning. Of the histopathological markers, only MAP 2 and SYP were associated with neurocognitive functioning; however, MAP 2 and SYP did not vary as a function of genotype. Mediation analysis suggests a causal pathway in which presynaptic degeneration (SYP) leads to somatodendritic degeneration (MAP 2) and ultimately neurocognitive impairment. This study did not support the role of host genotype in the histopathology underlying HAND. The findings lend further support for synaptodendritic degeneration as the proximal underlying neuropathological substrate of HAND.


Subject(s)
Cognitive Dysfunction/genetics , Dendrites/pathology , HIV Infections/genetics , Microtubule-Associated Proteins/genetics , Synapses/pathology , Synaptophysin/genetics , Adult , Cognitive Dysfunction/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cytokines/genetics , Dendrites/genetics , Dendrites/metabolism , Female , Frontal Lobe/metabolism , Frontal Lobe/pathology , Gene Expression , Genotype , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/pathology , Humans , Longitudinal Studies , Male , Microtubule-Associated Proteins/metabolism , Middle Aged , Neuropsychological Tests , Phenotype , Polymorphism, Genetic , Presynaptic Terminals/metabolism , Presynaptic Terminals/pathology , Synapses/genetics , Synapses/metabolism , Synaptophysin/metabolism
18.
Mol Psychiatry ; 24(4): 549-561, 2019 04.
Article in English | MEDLINE | ID: mdl-29511299

ABSTRACT

Although synaptic loss is thought to be core to the pathophysiology of schizophrenia, the nature, consistency and magnitude of synaptic protein and mRNA changes has not been systematically appraised. Our objective was thus to systematically review and meta-analyse findings. The entire PubMed database was searched for studies from inception date to the 1st of July 2017. We selected case-control postmortem studies in schizophrenia quantifying synaptic protein or mRNA levels in brain tissue. The difference in protein and mRNA levels between cases and controls was extracted and meta-analysis conducted. Among the results, we found a significant reduction in synaptophysin in schizophrenia in the hippocampus (effect size: -0.65, p < 0.01), frontal (effect size: -0.36, p = 0.04), and cingulate cortices (effect size: -0.54, p = 0.02), but no significant changes for synaptophysin in occipital and temporal cortices, and no changes for SNAP-25, PSD-95, VAMP, and syntaxin in frontal cortex. There were insufficient studies for meta-analysis of complexins, synapsins, rab3A and synaptotagmin and mRNA measures. Findings are summarised for these, which generally show reductions in SNAP-25, PSD-95, synapsin and rab3A protein levels in the hippocampus but inconsistency in other regions. Our findings of moderate-large reductions in synaptophysin in hippocampus and frontal cortical regions, and a tendency for reductions in other pre- and postsynaptic proteins in the hippocampus are consistent with models that implicate synaptic loss in schizophrenia. However, they also identify potential differences between regions and proteins, suggesting synaptic loss is not uniform in nature or extent.


Subject(s)
Schizophrenia/genetics , Schizophrenia/physiopathology , Synapses/genetics , Adult , Brain/metabolism , Case-Control Studies , Disks Large Homolog 4 Protein/metabolism , Female , Gyrus Cinguli/metabolism , Hippocampus/metabolism , Humans , Male , Middle Aged , RNA, Messenger/metabolism , Synapses/metabolism , Synapsins/metabolism , Synaptic Vesicles/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism , Synaptosomal-Associated Protein 25/metabolism , Temporal Lobe/metabolism , rab3A GTP-Binding Protein/metabolism
19.
Addict Biol ; 25(1): e12697, 2020 01.
Article in English | MEDLINE | ID: mdl-30585381

ABSTRACT

Methamphetamine (METH) causes memory changes, but the underlying mechanisms are poorly understood. Epigenetic mechanisms, including DNA methylation, can potentially cause synaptic changes in the brain. Oxytocin (OT) plays a central role in learning and memory, but little is known of the impact of OT on METH-associated memory changes. Here, we explored the role of OT in METH-induced epigenetic alterations that underlie spatial and cognitive memory changes. METH (2.0 mg/kg, i.p.) was administered to male C57BL/6 mice once every other day for 8 days. OT (2.5 µg, i.c.v.) or aCSF was given prior to METH. Spatial and cognitive memory were assessed. In Hip and PFC, synaptic structures and proteins were examined, levels of DNA methyltransferases (DNMTs) and methyl CpG binding protein 2 (MECP2) were determined, and the DNA methylation status at the Synaptophysin (Syn) promoter was assessed. METH enhanced spatial memory, decreased synapse length, downregulated DNMT1, DNMT3A, DNMT3B, and MECP2, and induced DNA hypomethylation at the Syn promoter in Hip. In contrast, METH reduced cognitive memory, increased synapse thickness, upregulated DNMT1, DNMT3A, and MECP2, and induced DNA hypermethylation at the Syn promoter in PFC. OT pretreatment specifically ameliorated METH-induced learning and memory alterations, normalized synapse structures, and regulated DNMTs and MECP2 to reverse the DNA methylation status changes at the Syn promoter in Hip and PFC. DNA methylation is an important gene regulatory mechanism underlying METH-induced learning and memory alterations. OT can potentially be used to specifically manipulate METH-related memory changes.


Subject(s)
Amphetamine-Related Disorders/physiopathology , DNA Methylation/drug effects , Learning/drug effects , Methamphetamine/pharmacology , Oxytocin/pharmacology , Synaptophysin/genetics , Amphetamine-Related Disorders/metabolism , Animals , Central Nervous System Stimulants/pharmacology , Disease Models, Animal , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Oxytocics/pharmacology , Promoter Regions, Genetic/drug effects , Synaptophysin/metabolism
20.
Mol Cell Neurosci ; 95: 86-95, 2019 03.
Article in English | MEDLINE | ID: mdl-30763689

ABSTRACT

The normal role of Alzheimer's disease (AD)-linked amyloid precursor protein (APP) in the brain remains incompletely understood. Previous studies have reported that lack of APP has detrimental effects on spines and electrophysiological parameters. APP has been described to be important in synaptic pruning during development. The effect of APP knockout on mature synapses is complicated by this role in development. We previously reported on differential changes in synaptic proteins and receptors in APP mutant AD transgenic compared to wild-type neurons, which revealed selective decreases in levels of pre- and post-synaptic proteins, including of surface glutamate receptors. In the present study, we undertook a similar analysis of synaptic composition but now in APP knockout compared to wild-type mouse neurons. Here we demonstrate alterations in levels of selective pre- and post-synaptic proteins and receptors in APP knockout compared to wild-type mouse primary neurons in culture and brains of mice in youth and adulthood. Remarkably, we demonstrate selective increases in levels of synaptic proteins, such as GluA1, in neurons with APP knockout and with RNAi knockdown, which tended to be opposite to the reductions seen in AD transgenic APP mutant compared to wild-type neurons. These data reinforce that APP is important for the normal composition of synapses.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Neurons/metabolism , Synapses/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cells, Cultured , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , Mice , Mice, Inbred C57BL , Neurons/cytology , Neuropeptides/genetics , Neuropeptides/metabolism , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL