VAMP1 Human

Synaptobrevin-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Isoforms

VAMP1 is a member of the SNARE (Soluble NSF Attachment Protein Receptor) family, which facilitates vesicle docking and fusion. Structurally, it consists of a conserved SNARE motif and a transmembrane domain. Alternative splicing generates multiple isoforms:

IsoformKey FeaturesExpressionSource
VAMP1AContains a hydrophobic C-terminal anchor; predominant in neuronal tissues.Brain, spinal cord
VAMP1BShortened hydrophobic anchor with charged C-terminal residues; targets mitochondria.Non-neuronal cells (e.g., endothelial cells)
VAMP1DLess characterized; potential tissue-specific roles.Under investigation

The mitochondrial localization of VAMP1B suggests a non-canonical role in organelle dynamics, potentially mediating mitochondrial fusion or inter-organelle communication .

Functional Role in Synaptic Transmission

VAMP1 is a core component of the SNARE complex, partnering with syntaxin and SNAP25 to catalyze membrane fusion . Key functions include:

  • Neurotransmitter Release: Mediates exocytosis of synaptic vesicles in motor neurons .

  • Synaptic Plasticity: Regulates synaptic strength and long-term potentiation .

  • Pathogen Interaction: Targeted by botulinum neurotoxins (BoNTs), which cleave VAMP1 to inhibit neurotransmitter release .

Clinical and Pathological Associations

VAMP1 mutations or dysregulation are implicated in several disorders:

DiseaseMutation/AssociationMechanismSource
Autosomal Dominant Spastic Ataxia 1Splice-site mutations disrupting VAMP1A expression.Loss of neuronal VAMP1 impairs synaptic transmission.
Congenital Myasthenic SyndromeHomozygous splicing variants (e.g., c.129+5 G>A, c.341-24_341-16delinsAGAAAA).Presynaptic neuromuscular junction dysfunction.
Alzheimer’s Disease (AD)SNPs in VAMP1 correlate with amyloid-beta (Aβ) production.Altered coupling of synaptic activity to Aβ generation.
Botulinum Toxin ResistancePolymorphism at residue 48 (I48/M48) determines sensitivity to BoNT/D in vertebrates.I48 in humans confers insensitivity to BoNT/D.

Knockout mouse models show reduced Aβ levels, suggesting VAMP1 as a therapeutic target for AD . Conversely, VAMP1 overexpression in lung cancer correlates with improved survival .

  • Evolutionary Adaptation: Frequent residue 48 polymorphisms (I/M) in primates suggest selective pressure from BoNTs .

  • Therapeutic Potential: Modulating VAMP1 expression or interaction partners could mitigate Aβ pathology in AD or enhance neuromuscular function in myasthenic syndromes .

Product Specs

Introduction
Synaptobrevin 1 (VAMP1), a vesicle-associated membrane protein, is crucial for regulated exocytosis as a key component of the SNARE complex. VAMP1 interacts with target SNAREs Syntaxin (STX) and SNAP25 following the fusion of synaptic vesicles with the plasma membrane.
Description
Recombinant Human VAMP1, expressed in E. coli, is a non-glycosylated polypeptide chain consisting of 111 amino acids (residues 1-91) with a molecular weight of 11.9 kDa. The N-terminus of VAMP1 is fused with a 20 amino acid His-Tag. Purification is achieved through standard chromatographic methods.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The protein solution is provided at a concentration of 1 mg/ml and is formulated in a buffer containing 1X PBS and 1 mM EDTA.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freeze-thaw cycles should be avoided.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Vesicle-associated membrane protein 1, SYB1, VAMP-1, Synaptobrevin-1, VAMP1, DKFZp686H12131.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MSAPAQPPAE GTEGTAPGGG PPGPPPNMTS NRRLQQTQAQVEEVVDIIRV NVDKVLERDQ KLSELDDRAD ALQAGASQFE SSAAKLKRKY W.

Q&A

What is VAMP1 and what is its primary function in human neuronal systems?

VAMP1, also known as synaptobrevin 1, functions as a critical component of the SNARE (Soluble N-ethylmaleimide-sensitive factor Attachment Protein REceptor) complex, which is essential for synaptic exocytosis in the human nervous system. This protein facilitates the fusion of synaptic vesicles with the presynaptic membrane, enabling neurotransmitter release.

Methodologically, VAMP1's function can be investigated through:

  • Biochemical assays examining protein-protein interactions

  • Electrophysiological recordings to measure synaptic transmission

  • Immunohistochemistry to determine localization patterns

VAMP1 shows specific expression patterns in the cerebellum, brainstem, and spinal cord, particularly in nuclei controlling eye movements, tongue movements, swallowing, and limb movements . This distribution pattern aligns with symptoms observed in VAMP1-associated neurological disorders, suggesting region-specific functions in neural circuits controlling motor functions.

How can researchers differentiate between VAMP1 isoforms in human tissues?

Human VAMP1 exists in several distinct isoforms with different tissue distributions and functional roles:

IsoformRefSeq IDPrimary ExpressionKey Features
VAMP1ANM_014231.3Nervous systemPrimary neuronal isoform
VAMP1BNM_016830.2Non-neuronal tissuesContains mitochondrial targeting signal
VAMP1DNM_199245.1Non-neuronal tissuesSimilar to rat Vamp1b

For accurate differentiation of these isoforms, researchers should employ:

  • RT-PCR with isoform-specific primers: Design primers spanning unique exon junctions or targeting isoform-specific regions

  • RNA-Seq analysis: Utilize computational approaches to distinguish between different isoform transcripts

  • Western blotting: Use antibodies recognizing unique C-terminal regions of each isoform

  • Immunohistochemistry: Employ isoform-specific antibodies for localization studies

RNA-Seq data from Illumina's Human BodyMap confirms the predominance of VAMP1A in brain tissue, with minimal expression of VAMP1B and no detectable VAMP1D in the brain . This mutual exclusivity in expression patterns is important for researchers to consider when designing experiments.

What methodologies are most effective for measuring VAMP1 expression in clinical samples?

When analyzing VAMP1 expression in clinical samples, researchers should consider a complementary approach:

  • Transcriptional Analysis:

    • qRT-PCR with isoform-specific primers

    • RNA-Seq for comprehensive transcriptome analysis

    • Digital droplet PCR for absolute quantification

  • Protein Detection:

    • Western blotting for protein levels

    • ELISA for quantitative measurement

    • Immunohistochemistry for spatial distribution

  • Genetic Analysis:

    • Genotyping of expression-modifying polymorphisms

    • Expression quantitative trait loci (eQTL) analysis

    • Analysis of splicing using minigene constructs

For correlating genotype with expression, research indicates that multiple polymorphisms within VAMP1 are associated with altered expression levels. For example, all 8 polymorphisms examined in one study showed unequivocal association with altered VAMP1 expression (p < 3.7×10^-4) . When analyzing cerebellar tissue, rs7390 showed strong association with increased VAMP1 expression, providing a potential genetic marker for expression levels .

What is the relationship between VAMP1 and other components of the SNARE complex?

VAMP1 functions within the SNARE complex through specific protein-protein interactions:

SNARE ComponentSubcellular LocationInteraction with VAMP1Functional Role
Syntaxin-1Plasma membraneForms core SNARE complexDocking/fusion
SNAP-25Plasma membraneForms core SNARE complexDocking/fusion
SynaptotagminVesicle membraneCalcium-dependent interactionFusion trigger
NSF/α-SNAPCytoplasmicDisassembly of SNARE complexRecycling

To investigate these interactions, researchers can employ:

  • Co-immunoprecipitation assays

  • FRET (Förster Resonance Energy Transfer) analysis

  • In vitro reconstitution of membrane fusion

  • Crystallography of protein complexes

Understanding these interactions is particularly important when investigating how VAMP1 mutations might disrupt normal synaptic transmission in neurological disorders like hereditary spastic ataxia .

What are the known genetic variants of VAMP1 and their functional consequences?

VAMP1 exhibits both common polymorphisms and rare variants with distinct functional impacts:

Common Variants:

  • rs2072376: Associated with decreased cerebellar VAMP1 expression and potentially protective against Alzheimer's disease (OR = 0.88, p = 0.03)

  • rs7390: Associated with increased VAMP1 expression

Rare Variants:

  • rs74056956: Associated with increased Alzheimer's disease risk (OR = 2.11, p = 0.05)

  • rs71584834: Associated with increased Alzheimer's disease risk (OR = 1.91, p = 0.0006)

  • Splice-site mutations affecting the donor site at exon 4 (c.340+2): Cause hereditary spastic ataxia through loss of the neuronal VAMP1A isoform

For functional characterization, researchers can use:

  • Minigene splicing assays to assess effects on RNA processing

  • Expression studies in neuronal cultures

  • Electrophysiological recordings to measure effects on neurotransmission

  • Animal or cellular models expressing variant forms

The functional impact of these variants appears to be isoform-specific, with mutations affecting the splicing donor site primarily disrupting the neuronal VAMP1A isoform .

How do mutations in VAMP1 lead to hereditary spastic ataxia?

Mutations in VAMP1 cause autosomal dominant hereditary spastic ataxia (SPAX1) through a haploinsufficiency mechanism affecting the neuron-specific VAMP1A isoform. The pathophysiological cascade involves:

  • Molecular Defect: A mutation affecting the critical donor site for splicing of VAMP1 isoforms (c.340+2) disrupts normal RNA processing

  • Isoform Impact: Loss of the VAMP1A isoform, which is the only isoform expressed in the nervous system

  • Functional Consequence: Reduced neurotransmitter exocytosis in specific brain regions

  • Anatomical Correlation: Effects on cerebellum, brainstem, and spinal cord align with observed symptoms

The clinical manifestations include:

  • Cerebellar ataxia

  • Spastic paraplegia

  • Supranuclear gaze palsy

  • Memory impairment

  • Dysphagia

  • Ptosis

These symptoms correlate with VAMP1's known expression pattern in nuclei controlling eye movements, tongue movements, swallowing, and limb movements . The variable phenotype seen in affected individuals suggests that VAMP1 should be considered in the differential diagnosis of patients with either ataxia or spastic paraplegia, particularly when both symptom complexes are present.

What is the relationship between VAMP1 expression and Alzheimer's disease pathophysiology?

Research has revealed a complex relationship between VAMP1 and Alzheimer's disease (AD) that involves both genetic association and functional interactions with amyloid processing:

  • Genetic Associations:

    • SNPs associated with increased VAMP1 expression tend to increase AD risk

    • SNPs associated with decreased VAMP1 expression may have a protective effect

    • The polymorphism rs2072376 is associated with decreased VAMP1 expression and showed a modest protective effect (OR = 0.88, p = 0.03)

  • Mechanistic Relationship:

    • VAMP1 appears to function as a coupling protein between vesicular release and APP processing

    • Variations in VAMP1 levels alter synaptic activity and consequently affect Aβ production

    • In VAMP1 knockout mice, endogenous Aβ40 and Aβ42 levels are substantially reduced

  • Research Methodology:

    • Case-control genetic association studies

    • eQTL analysis in brain tissue

    • Cellular models examining APP processing with varied VAMP1 expression

    • Measurement of Aβ species in relation to VAMP1 levels

The current hypothesis suggests that VAMP1-mediated synaptic activity influences APP processing by altering the rate of vesicle endocytosis, which is critical for APP processing into Aβ . This provides a potential mechanistic link between synaptic activity and AD pathogenesis that warrants further investigation.

What experimental models best replicate human VAMP1 function in disease states?

Selecting appropriate experimental models for VAMP1 research requires careful consideration of their advantages and limitations:

Model SystemAdvantagesLimitationsBest Applications
Human iPSC-derived neuronsHuman genetic background; patient-specific mutationsMaturation time; variabilityPatient-specific disease modeling; drug screening
VAMP1 knockout/knock-in miceIn vivo system; behavioral assessmentSpecies differences in isoform expressionSystemic effects; long-term consequences
Primary neuronal culturesControlled environment; easy manipulationLimited lifespan; altered networkMechanistic studies; electrophysiology
Brain organoids3D architecture; multiple cell typesLack of vascularization; variabilityDevelopmental aspects; cell-cell interactions
In vitro reconstitutionPrecise control; direct measurementSimplified systemBiophysical properties of membrane fusion

For studying hereditary spastic ataxia, humanized mouse models carrying patient-specific mutations would be valuable, though the existing lethal wasting (lew/lew) mouse model has limitations due to its severe phenotype .

For Alzheimer's disease research, iPSC-derived neurons expressing different VAMP1 variants combined with Aβ measurements provide a system for studying how VAMP1 influences APP processing . The selection of the appropriate model should align with the specific research question regarding VAMP1 function or dysfunction.

How do VAMP1 isoforms differ functionally in human synaptic transmission?

The functional differences between VAMP1 isoforms have significant implications for synaptic physiology:

IsoformSubcellular LocalizationFunctional RoleExpression Pattern
VAMP1ASynaptic vesiclesMediates neurotransmitter releaseCerebellum, brainstem, spinal cord
VAMP1BMitochondria in non-neuronal cellsPotentially involved in mitochondrial functionBroad non-neuronal expression
VAMP1DNon-neuronal tissuesPoorly characterizedNot expressed in nervous system

These isoforms differ primarily in their C-terminal regions, with VAMP1B containing a mitochondrial targeting signal not present in VAMP1A . The mutually exclusive expression pattern suggests distinct evolutionary adaptations for specialized functions.

The importance of isoform specificity is highlighted in disease states:

  • Mutations affecting the splicing donor site at exon 4 (c.340+2) primarily affect VAMP1A production

  • Loss of VAMP1A in neurons leads to hereditary spastic ataxia

  • VAMP1B, being absent in neurons, cannot compensate for VAMP1A deficiency

For studying isoform-specific functions, researchers should employ:

  • Selective knockdown/overexpression of specific isoforms

  • Electrophysiological recordings to assess effects on transmission

  • Live imaging with isoform-specific tags to track localization

  • Proteomics to identify isoform-specific interaction partners

What methodological approaches should be used to study the relationship between VAMP1 polymorphisms and APP processing?

Investigating how VAMP1 polymorphisms affect APP processing and Aβ production requires sophisticated methodological approaches:

  • Genetic Association Studies:

    • Case-control studies correlating VAMP1 variants with AD risk

    • Family-based association studies for rare variants

    • Genome-wide interaction studies to identify modifiers

  • Expression Analysis:

    • eQTL analysis to link variants to expression levels

    • Allele-specific expression to detect cis-regulatory effects

    • Single-cell RNA-seq to identify cell-type specific effects

  • Functional Studies:

    • Neuronal cultures expressing different VAMP1 variants

    • CRISPR-engineered isogenic lines with specific variants

    • Measurement of synaptic vesicle release using FM dyes or pHluorins

    • Quantification of APP processing products (Aβ40, Aβ42) via ELISA

    • Live imaging of APP trafficking with fluorescent tags

  • Systems Biology Approaches:

    • Pathway analysis integrating VAMP1 with APP processing networks

    • Computational modeling of how altered vesicle dynamics affect APP processing

    • Multi-omics integration to identify disease signatures

Current evidence suggests that VAMP1 functions as a coupling protein between vesicular release and APP processing, with variations in VAMP1 levels altering Aβ production . The substantial reduction of endogenous Aβ in VAMP1 knockout mice provides compelling evidence for this relationship.

What are the clinical and research implications of VAMP1-associated symptoms for neurological diagnostics?

VAMP1 mutations and variants have significant implications for clinical practice and research in neurology:

Clinical Implications:

  • Diagnostic Considerations:

    • VAMP1 should be tested in patients with combined ataxia and spastic paraplegia

    • Patients with ancestral links to Newfoundland (Canada) have higher risk for the founder mutation

    • Common symptoms include ptosis (80-100%), dysphagia (80-100%), pes cavus (80-100%), seizures (50-80%), and leg muscle stiffness (50-80%)

  • Phenotypic Spectrum:

    • Additional common symptoms include ataxia, supranuclear gaze palsy, slow saccadic eye movements, spastic gait, memory impairment, and difficulty walking

    • The variable phenotypic presentation necessitates broad clinical awareness

Research Implications:

  • Genotype-Phenotype Correlations:

    • Different mutations may affect specific isoforms differently

    • Position of mutations within functional domains may predict symptom severity

    • Modifier genes may explain phenotypic variability

  • Biomarker Development:

    • VAMP1 expression levels as potential biomarkers for disease progression

    • Isoform ratios as indicators of disease state

    • Association with other synaptic markers for comprehensive profiling

  • Therapeutic Targeting:

    • Isoform-specific interventions may provide precision medicine approaches

    • Gene therapy strategies to restore VAMP1A function in hereditary spastic ataxia

    • Modulation of VAMP1 levels as a potential approach in Alzheimer's disease

The identification of VAMP1 mutations in neurological disorders highlights the importance of proteins involved in membrane-trafficking and axonal transport, connecting VAMP1 to broader families of movement disorders and neurodegenerative conditions .

How can VAMP1 be targeted therapeutically in different neurological disorders?

Therapeutic strategies targeting VAMP1 must be tailored to the specific pathophysiology of each disorder:

For Hereditary Spastic Ataxia (VAMP1 Haploinsufficiency):

  • Gene Augmentation Approaches:

    • AAV-mediated delivery of functional VAMP1A to affected neurons

    • Cell-type specific promoters to target highly affected regions

    • Antisense oligonucleotides to correct splicing defects

  • Enhancement of Remaining Function:

    • Small molecules that enhance SNARE complex assembly

    • Compounds that increase the efficiency of remaining VAMP1

    • Stabilization of VAMP1 protein to increase functional half-life

For Alzheimer's Disease (Where Increased VAMP1 May Contribute):

  • Modulation of VAMP1 Expression:

    • Targeted reduction of VAMP1 levels to reduce Aβ production

    • Compounds that modify APP-VAMP1 interactions

    • Regulators of VAMP1 transcription or translation

  • Alteration of VAMP1 Function:

    • Compounds that modify VAMP1's role in vesicle fusion without blocking neurotransmission

    • Selective modulators of VAMP1 in amyloidogenic pathways

    • Targeting VAMP1 post-translational modifications

Methodological Considerations:

  • Target Validation:

    • Knockdown/overexpression studies in relevant models

    • Structure-function analysis to identify druggable domains

    • Temporal control systems to determine intervention windows

  • Delivery Challenges:

    • Blood-brain barrier penetration strategies

    • Cell-type specific targeting to minimize off-target effects

    • Sustained vs. pulsatile delivery based on disease mechanisms

The critical involvement of VAMP1 in basic neurotransmission presents a significant challenge, requiring therapeutic strategies that modulate pathological processes while preserving essential synaptic functions.

How do research findings on VAMP1 connect to other neurological disorders beyond hereditary spastic ataxia and Alzheimer's disease?

VAMP1's fundamental role in synaptic transmission suggests potential involvement in multiple neurological conditions:

  • Other Movement Disorders:

    • VAMP1's expression in cerebellum and motor pathways suggests possible roles in additional ataxias

    • The protein's function in vesicular release connects to broader synaptic dysfunction in movement disorders

    • Research methodologies should include VAMP1 analysis in unexplained movement disorder cohorts

  • Epilepsy:

    • Seizures are reported in 50-80% of patients with VAMP1 mutations

    • Altered neurotransmitter release due to VAMP1 dysfunction may contribute to abnormal circuit excitability

    • VAMP1 should be included in epilepsy gene panels, particularly for cases with ataxia or spasticity

  • Synaptic Plasticity Disorders:

    • VAMP1's role in vesicle release impacts long-term potentiation and depression

    • Cognitive symptoms in patients with VAMP1 mutations suggest broader effects on plasticity

    • Research should examine VAMP1 variants in learning and memory disorders

  • Other Neurodegenerative Conditions:

    • VAMP1 belongs to the membrane-trafficking and axonal transport family of proteins

    • This protein family has been linked to both dominant ataxias and hereditary spastic paraplegias

    • VAMP1 interaction networks overlap with other neurodegenerative disease pathways

Research approaches connecting VAMP1 to broader neurological contexts should include:

  • Network analysis of VAMP1 interactors across disease proteomes

  • Model systems examining VAMP1 in diverse neuronal populations

  • Population genetics to identify VAMP1 variants enriched in neurological conditions

The diverse symptomatology of confirmed VAMP1-related disorders suggests this protein warrants investigation across a broader spectrum of neurological conditions.

What are the current challenges and future directions in VAMP1 research?

Several key challenges and opportunities exist in the field of VAMP1 research:

Current Challenges:

  • Isoform-Specific Functions:

    • Limited tools for distinguishing isoforms in vivo

    • Difficulty in selectively manipulating specific isoforms

    • Incomplete understanding of isoform switching in development and disease

  • Rare Variant Characterization:

    • Low frequency makes functional studies challenging

    • Heterozygous effects difficult to model in vitro

    • Variants of uncertain significance require functional validation

  • Tissue-Specific Regulation:

    • Mechanisms controlling differential splicing poorly understood

    • Cell-type specific expression patterns need better characterization

    • Regulatory elements controlling VAMP1 expression not fully mapped

Future Directions:

  • Advanced Methodologies:

    • Single-cell multi-omics to map VAMP1 isoform expression at cellular resolution

    • CRISPR-based approaches for isoform-specific manipulation

    • Improved structural biology techniques to understand VAMP1 conformational dynamics

  • Translational Opportunities:

    • Development of isoform-specific biomarkers for neurological disorders

    • Gene therapy approaches for VAMP1-associated diseases

    • Drug discovery targeting VAMP1 interactions or expression

  • Integrative Approaches:

    • Systems biology integration of VAMP1 into synaptic function networks

    • Population-scale analysis of VAMP1 variation and neurological phenotypes

    • Multi-modal imaging to connect VAMP1 dysfunction to circuit-level abnormalities

The continued investigation of VAMP1's role in both normal synaptic function and disease states holds promise for advances in understanding fundamental neurobiology and developing new therapeutic approaches for neurological disorders.

Product Science Overview

Introduction

Synaptobrevin-1, also known as VAMP1 (Vesicle-Associated Membrane Protein 1), is a crucial protein involved in the process of synaptic vesicle fusion and neurotransmitter release in neurons. It is a member of the SNARE (Soluble NSF Attachment Protein Receptor) protein family, which plays a vital role in the fusion of vesicles with target membranes, a process essential for neurotransmission.

Structure

Synaptobrevin-1 is a small integral membrane protein with a molecular weight of approximately 18 kilodaltons (kDa). It is characterized by a single transmembrane domain and a cytoplasmic domain that participates in the formation of the SNARE complex. The SNARE complex is composed of four α-helices: one contributed by synaptobrevin, one by syntaxin, and two by SNAP-25 .

Function

The primary function of synaptobrevin-1 is to mediate the fusion of synaptic vesicles with the presynaptic membrane, facilitating the release of neurotransmitters into the synaptic cleft. This process is critical for the transmission of signals between neurons. Synaptobrevin-1, along with other SNARE proteins, forms a tight complex that brings the vesicle and target membranes into close proximity, allowing them to fuse .

Role in Neurotransmission

Synaptobrevin-1 is essential for the proper functioning of the nervous system. It is involved in the regulated release of neurotransmitters, which is crucial for various physiological processes, including memory consolidation, mood regulation, and overall neuronal communication. Disruptions in synaptobrevin-1 function can lead to neurological disorders such as epilepsy, depression, and neurodegeneration .

Clinical Significance

Synaptobrevin-1 is a target for various bacterial toxins, including tetanospasmin (produced by Clostridium tetani) and botulinum toxin (produced by Clostridium botulinum). These toxins cleave synaptobrevin-1, inhibiting neurotransmitter release and leading to conditions such as tetanus and botulism .

Recombinant Synaptobrevin-1

Recombinant synaptobrevin-1 is produced using genetic engineering techniques, where the gene encoding synaptobrevin-1 is inserted into an expression system, such as bacteria or yeast, to produce the protein in large quantities. This recombinant protein is used in various research applications to study the mechanisms of synaptic vesicle fusion and neurotransmitter release.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.