SYT11 Human

Synaptotagmin XI Human Recombinant
Shipped with Ice Packs
In Stock

Description

Parkinson’s Disease (PD)

  • Pathogenic Mechanism: SYT11 accumulation due to parkin mutations inhibits dopamine (DA) vesicle endocytosis, impairing striatal DA release and causing neurodegeneration .

  • Key Interaction: SYT11 is ubiquitinated and degraded by parkin (E3 ligase). PD-linked parkin mutants (e.g., R42P, R275W) fail to regulate SYT11, leading to neurotoxicity .

  • Palmitoylation: Post-translational modification at Cys39/40 stabilizes SYT11, promoting α-synuclein aggregation—a hallmark of PD .

Schizophrenia

  • Genetic Risk: Reduced SYT11 expression in the prefrontal cortex and plasma correlates with schizophrenia. Plasma SYT11 levels show diagnostic potential (AUC = 0.737) .

  • Behavioral Deficits: DA neuron-specific SYT11 knockout (cKO) mice exhibit social interaction deficits and hyperlocomotion, mimicking schizophrenia symptoms .

Synaptic Regulation

  • Endocytosis Inhibition: SYT11 limits clathrin-mediated and bulk endocytosis, ensuring precision in vesicle retrieval .

  • Vesicle Trafficking: Facilitates GABA<sub>B</sub>/Cav2.2 complex assembly in post-Golgi vesicles, stabilizing presynaptic Ca²⁺ channels and G protein-coupled receptors .

In Vitro and Preclinical Studies

  • Neurotoxicity Model: Overexpression of full-length SYT11 in murine substantia nigra causes DA neuron degeneration and motor deficits, reversible by SYT11 knockdown .

  • Knockout Phenotypes:

    • Global SYT11 KO in mice results in neonatal lethality due to feeding defects .

    • Forebrain-specific cKO mice show spatial memory deficits but intact fear memory .

Interaction Network and Pathways

SYT11 interacts with:

  • Parkin: Mediates ubiquitination and degradation of SYT11 .

  • KCTD16/Cav2.2: Forms presynaptic signaling complexes to regulate neurotransmitter release .

  • Clathrin/Dynamin: Modulates endocytosis kinetics by restricting membrane invagination .

Clinical and Therapeutic Implications

DiseaseSYT11 RoleTherapeutic Target
Parkinson’s DiseaseAccumulation disrupts vesicle recyclingInhibit palmitoylation or enhance parkin activity
SchizophreniaDeficiency alters DA transmissionSYT11 augmentation or gene therapy

Product Specs

Introduction
Synaptotagmin-11 (SYT11) is a protein that potentially plays a role in the calcium-dependent release of substances from secretory vesicles. It might achieve this through binding to calcium ions and phospholipids via its C2 domain. Additionally, SYT11 may act as a calcium sensor during vesicle trafficking and exocytosis.
Description
This product consists of a single, non-glycosylated polypeptide chain of the human SYT11 protein, containing 418 amino acids (residues 37-431) with a molecular weight of 47kDa. It is produced through recombinant expression in E. coli. The protein includes a 23 amino acid His-tag at the N-terminus to facilitate purification, which is carried out using proprietary chromatographic methods.
Physical Appearance
The product appears as a clear, colorless solution that has been sterilized through filtration.
Formulation
This SYT11 protein solution has a concentration of 0.25mg/ml and is supplied in a buffer consisting of Phosphate Buffered Saline (pH 7.4) and 50% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. Adding a carrier protein such as 0.1% HSA or BSA is advisable for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity
The purity of the SYT11 protein in this product is greater than 80%, as determined by SDS-PAGE analysis.
Synonyms
Synaptotagmin XI, SytXI, Synaptotagmin 12, KIAA0080, SYT12, Synaptotagmin-11.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSWSCCHQQ AEKKQKNPPY KFIHMLKGIS IYPETLSNKK KIIKVRRDKD GPGREGGRRN LLVDAAEAGL LSRDKDPRGP SSGSCIDQLP IKMDYGEELR SPITSLTPGE SKTTSPSSPE EDVMLGSLTF SVDYNFPKKA LVVTIQEAHG LPVMDDQTQG SDPYIKMTIL PDKRHRVKTR VLRKTLDPVF DETFTFYGIP YSQLQDLVLH FLVLSFDRFS RDDVIGEVMV PLAGVDPSTG KVQLTRDIIK RNIQKCISRG ELQVSLSYQP VAQRMTVVVL KARHLPKMDI TGLSGNPYVK VNVYYGRKRI AKKKTHVKKC TLNPIFNESF IYDIPTDLLP DISIEFLVID FDRTTKNEVV GRLILGAHSV TASGAEHWRE VCESPRKPVA KWHSLSEY.

Q&A

What is SYT11 and what are its primary functions in the human nervous system?

Synaptotagmin-11 (SYT11) is a non-Ca²⁺-binding member of the synaptotagmin family that functions as a key regulator of neurotransmission in the central nervous system. Unlike other synaptotagmins that primarily mediate calcium-dependent exocytosis, SYT11 serves as an inhibitor of spontaneous release by interacting with vps10p-tail-interactor-1a (vti1a), a non-canonical SNARE protein . It plays critical roles in regulating dopamine transmission, facilitating the assembly of presynaptic signaling complexes, and modulating vesicle trafficking. Specifically, SYT11 functions as an integral vesicle protein that inhibits various forms of endocytosis, including clathrin-mediated and bulk endocytosis in neurons . Additionally, it facilitates the assembly of GABAB receptor/Cav2.2 signaling complexes by recruiting both components to post-Golgi transport vesicles, ultimately influencing neurotransmitter release .

How does SYT11 expression vary across different brain regions and developmental stages?

SYT11 expression shows regional specificity and developmental regulation in the brain. Research has demonstrated that SYT11 is particularly significant in dopaminergic neurons of the substantia nigra pars compacta (SNpc), where alterations in its expression levels are associated with neurodegenerative processes . Notably, the timing of SYT11 deficiency appears critical for phenotype development; studies have shown that SYT11 deficiency in dopamine neurons during early adolescence, but not in adulthood, leads to persistent social deficits and schizophrenia-like behaviors in mouse models . This temporal specificity suggests differential expression and functional importance of SYT11 across developmental stages. Expression also extends beyond neurons to microglia, where SYT11 regulates immune functions including cytokine release and phagocytosis, indicating its diverse roles across different cell types in the brain .

What experimental methods are commonly used to measure SYT11 expression levels in brain tissue?

Several methodological approaches are employed to quantify SYT11 expression in brain tissue:

  • Western blot analysis: This technique allows for quantitative comparison of SYT11 protein levels between different brain regions or experimental conditions. For instance, researchers have used fluorescence-activated cell sorting (FACS) to collect transfected neurons followed by Western blot analysis to measure endogenous SYT11 expression changes in response to parkin overexpression .

  • Immunohistochemistry: This approach enables visualization of SYT11 distribution within specific brain structures. Studies have employed this method to examine Syt11 levels in the substantia nigra following viral-mediated gene knockdown, allowing for ipsilateral/contralateral comparison within the same animal .

  • Quantitative PCR (qPCR): Used to measure SYT11 mRNA levels, this technique is valuable for assessing transcriptional regulation.

  • Viral-mediated gene manipulation: Researchers frequently use lentiviral vectors expressing SYT11 shRNA or overexpression constructs to modulate SYT11 levels in specific brain regions, followed by molecular or behavioral assessments .

How is SYT11 linked to schizophrenia pathophysiology?

SYT11 has emerged as a potential genetic risk factor for schizophrenia through multiple lines of evidence. Recent research has identified reduced SYT11 expression in individuals with schizophrenia, with expression levels restored following antipsychotic treatment . Mechanistically, SYT11 deficiency specifically in dopamine neurons leads to dopamine over-transmission, which appears to be a key pathophysiological process. When this deficiency occurs during early adolescence (but not adulthood), it triggers persistent social deficits and other schizophrenia-like behaviors in mouse models .

The molecular pathway involves dopamine neuron over-excitation before late adolescence, leading to structural and functional alterations in the medial prefrontal cortex (mPFC). Importantly, interventions targeting dopamine D2 receptors (D2R) either presynaptically or postsynaptically demonstrate both acute and long-lasting therapeutic effects on social deficits in these schizophrenia mouse models . This suggests SYT11's role in schizophrenia occurs through its regulation of dopaminergic neurotransmission, particularly during critical developmental periods.

What is the relationship between SYT11, parkin, and Parkinson's disease pathogenesis?

SYT11 has been identified as a physiological substrate of parkin, an E3 ubiquitin ligase whose loss-of-function mutations represent the most common cause of autosomal recessive Parkinson's disease (PD). Research has demonstrated that parkin regulates SYT11 protein levels through ubiquitin-dependent proteasome degradation . In parkin-deficient models, SYT11 accumulates abnormally, leading to PD-like neurotoxicity.

The pathogenic mechanism involves several key steps:

  • SYT11 accumulation in the substantia nigra pars compacta impairs striatal dopamine release

  • This accumulation causes late-onset degeneration of dopaminergic neurons

  • Progressive contralateral motor abnormalities develop as a consequence

Critically, experimental manipulation of SYT11 levels can reverse parkin-associated pathology. When SYT11 is knocked down in the SNpc or specifically knocked out in dopaminergic neurons, the PD-like neurotoxicity induced by parkin dysfunction is significantly ameliorated . Mechanistically, SYT11 impairs vesicle pool replenishment and dopamine release by inhibiting endocytosis, making it a critical mediator of the neurodegenerative process in parkin-linked PD.

How do experimental models of SYT11 dysregulation recapitulate neuropsychiatric disease phenotypes?

Experimental models manipulating SYT11 expression have successfully recapitulated several key aspects of neuropsychiatric disorders:

  • Schizophrenia models:

    • Conditional knockout of SYT11 specifically in dopamine neurons during early adolescence produces persistent social deficits reminiscent of negative symptoms in schizophrenia

    • These models show dopamine over-transmission, consistent with the dopamine hypothesis of schizophrenia

    • Structural and functional alterations in the mPFC mirror those observed in schizophrenia

  • Parkinson's disease models:

    • Unilateral overexpression of full-length (but not C2B-truncated) SYT11 in the SNpc impairs ipsilateral striatal dopamine release

    • These models demonstrate late-onset degeneration of dopaminergic neurons

    • Progressive contralateral motor abnormalities develop, mimicking the motor symptoms of PD

    • Amperometric recordings with electrochemical carbon fiber electrodes (CFEs) in striatal slices show significantly reduced dopamine release in SYT11-overexpressing animals

  • Molecular phenotypes:

    • SYT11 manipulation in microglia affects cytokine production and phagocytosis, potentially modeling neuroinflammatory aspects of neuropsychiatric disorders

How does SYT11 regulate spontaneous neurotransmitter release?

SYT11 functions as a key regulator of spontaneous neurotransmitter release through its interaction with the non-canonical SNARE protein vps10p-tail-interactor-1a (vti1a). Experimental evidence from knockout and overexpression studies demonstrates that SYT11 acts as a suppressor of spontaneous release. In SYT11-knockout hippocampal neurons, researchers observed a significant increase in the frequency of miniature excitatory post-synaptic currents (mEPSCs), while overexpression of SYT11 resulted in decreased mEPSC frequency . Importantly, neither manipulation affected the average amplitude of these events, indicating that SYT11 exerts its effect through presynaptic regulation rather than postsynaptic mechanisms.

The molecular interaction between SYT11 and vti1a has been extensively characterized through multiple complementary approaches:

  • Glutathione S-transferase pull-down assays

  • Co-immunoprecipitation experiments

  • Affinity-purification methods

These techniques have confirmed a direct physical interaction between SYT11 and vti1a. The functional significance of this interaction was validated by the observation that knockdown of vti1a reversed the phenotype of elevated spontaneous release in SYT11 knockout neurons, identifying vti1a as the primary target of SYT11 inhibition . Domain analysis revealed that the C2A domain of SYT11 binds vti1a with high affinity, and expression of this domain alone is sufficient to rescue the phenotype in SYT11-knockout neurons, comparable to the effect of the full-length protein .

What protein-protein interactions mediate SYT11 function in vesicle trafficking?

SYT11 engages in several critical protein-protein interactions that collectively mediate its function in vesicle trafficking:

  • Interaction with SNARE proteins:

    • SYT11 directly binds to vti1a and vti1b through its linker domain, as demonstrated by biochemical interaction studies

    • This interaction is functionally significant for regulating spontaneous neurotransmission and cytokine release

  • Interaction with presynaptic signaling complexes:

    • SYT11 facilitates the assembly of GABAB receptor/Cav2.2 signaling complexes by recruiting both components to post-Golgi transport vesicles

    • This facilitatory role is essential for the proper delivery of these components to the presynaptic membrane

  • Interaction with endocytic machinery:

    • SYT11 inhibits endocytosis of GABAB receptors and Cav2.2 channels from the plasma membrane

    • SYT11 knockout mice exhibit a deficit in presynaptic Cav2.2 channels and GABAB receptors

    • These interactions collectively impact both neurotransmitter release and GABAB receptor-mediated inhibition of neurotransmitter release

  • Interaction with parkin:

    • SYT11 is a substrate for parkin, which regulates SYT11 levels through ubiquitin-dependent proteasome degradation

    • This interaction has significant implications for Parkinson's disease pathophysiology

What are the structural determinants of SYT11 function compared to other synaptotagmin family members?

SYT11 possesses distinctive structural features that differentiate it from other synaptotagmin family members and determine its unique functional properties:

  • Non-Ca²⁺-binding C2 domains:

    • Unlike canonical synaptotagmins (Syt1-7), SYT11 belongs to the subfamily of non-Ca²⁺-binding synaptotagmins

    • Its C2 domains lack the critical aspartate residues required for Ca²⁺ coordination, resulting in Ca²⁺-independent function

  • Domain-specific functions:

    • The C2A domain of SYT11 binds vti1a with high affinity and is sufficient to suppress spontaneous neurotransmission

    • The C2B domain appears to be crucial for dopamine release and neurotoxicity, as C2B-truncated SYT11 fails to induce the same effects as full-length SYT11 in vivo

  • Linker region importance:

    • The linker domain of SYT11 interacts with vti1a/vti1b, and peptides derived from this region can competitively inhibit this interaction

    • When applied to wild-type microglia, these peptides phenocopy the defects observed in SYT11 knockdown cells, inducing increased cytokine secretion upon LPS treatment

  • Transmembrane domain:

    • Like other synaptotagmins, SYT11 is an integral membrane protein with a single transmembrane domain that anchors it to vesicles

    • This localization is essential for its function in regulating vesicle trafficking and inhibiting endocytosis

What are the optimal methods for conditional manipulation of SYT11 expression in specific neural populations?

Several sophisticated approaches can be employed for conditional manipulation of SYT11 expression in specific neural populations:

  • Cre-loxP conditional knockout systems:

    • Researchers have successfully developed inducible microglia-specific SYT11-conditional-knockout (cKO) mouse strains to study SYT11 function in vivo

    • For dopaminergic neuron-specific manipulation, a similar approach can be combined with tyrosine hydroxylase (TH) promoter-driven Cre expression

    • These systems allow for temporal control when using tamoxifen-inducible Cre recombinase (CreERT2)

  • Viral-mediated gene delivery:

    • AAV vectors expressing SYT11 or shRNA against SYT11 under cell-type-specific promoters can achieve localized manipulation

    • Unilateral injection methods allow for powerful within-subject control designs, comparing ipsilateral and contralateral effects

    • For example, researchers have successfully used unilateral overexpression of SYT11 in the substantia nigra pars compacta to study its effects on dopamine release and motor behavior

  • CRISPR-Cas9 approaches:

    • In vivo CRISPR-Cas9 editing can be employed for targeted manipulation of the SYT11 gene in specific cell populations

    • This approach allows for precise modification of specific domains to study structure-function relationships

  • Chemogenetic and optogenetic integration:

    • Combining SYT11 manipulation with DREADD (Designer Receptors Exclusively Activated by Designer Drugs) or optogenetic tools enables investigation of circuit-specific effects of SYT11 dysregulation

How can researchers effectively measure the impact of SYT11 on neurotransmitter release dynamics?

Advanced techniques for measuring SYT11's impact on neurotransmitter release include:

  • Amperometric recordings with electrochemical carbon fiber electrodes (CFEs):

    • This technique has been successfully employed in striatal slices to quantify dopamine release following SYT11 manipulation in the substantia nigra

    • When local electrical stimulation is applied to striatal slices, CFEs can detect transient increases in amperometric current corresponding to dopamine release

    • Studies have shown that unilateral overexpression of SYT11 in the SNpc markedly decreases dopamine release in the ipsilateral striatum compared to the contralateral side

  • Whole-cell patch-clamp electrophysiology:

    • Recording miniature excitatory post-synaptic currents (mEPSCs) provides insights into spontaneous neurotransmitter release

    • SYT11 knockout hippocampal neurons show increased mEPSC frequency while SYT11 overexpression decreases it, without affecting amplitude

  • Optical imaging with genetically-encoded sensors:

    • Fluorescent reporters like GCaMP (for calcium) or iGluSnFR (for glutamate) can visualize neurotransmission dynamics

    • These approaches can be combined with SYT11 manipulation to assess effects on release probability and kinetics

  • Total Internal Reflection Fluorescence (TIRF) microscopy:

    • This technique can visualize single vesicle fusion events in cultured neurons

    • By combining TIRF with fluorescently tagged SYT11 and other vesicular proteins, researchers can track the spatiotemporal dynamics of SYT11-containing vesicles

What approaches can resolve contradictory findings regarding SYT11 function across different experimental models?

To address contradictory findings in SYT11 research, several methodological approaches are recommended:

  • Standardized experimental conditions:

    • Age-dependent effects are critical; SYT11 deficiency in dopamine neurons during early adolescence, but not adulthood, leads to persistent behavioral deficits

    • Therefore, precise developmental timing of manipulations should be standardized across studies

  • Cell-type specific analysis:

    • SYT11 functions differently in various cell types (neurons vs. microglia, dopaminergic vs. glutamatergic neurons)

    • Utilizing cell-type-specific manipulations and readouts can resolve seemingly contradictory findings

    • For example, using inducible microglia-specific SYT11-conditional-knockout mice revealed microglia-specific functions that differ from neuronal roles

  • Domain-specific functional analysis:

    • Different domains of SYT11 mediate distinct functions

    • The C2A domain binds vti1a and regulates spontaneous release

    • Full-length SYT11, but not C2B-truncated SYT11, impairs dopamine release and induces neurodegeneration

    • Domain-specific mutations or truncations can help dissect contradictory findings

  • Comprehensive phenotyping approach:

    • Employing multiple complementary techniques to assess SYT11 function:

      • Biochemical (protein-protein interactions)

      • Electrophysiological (neurotransmission)

      • Behavioral (motor function, cognition)

      • Anatomical (neurodegeneration)

    • This multilevel analysis can identify where contradictions arise and reconcile seemingly disparate findings

How might SYT11-targeted interventions be developed for neuropsychiatric disorders?

Based on current research, several promising approaches for SYT11-targeted interventions include:

  • D2 receptor modulation in schizophrenia:

    • Research has demonstrated that local intervention of D2R with clinical drugs, either presynaptically or postsynaptically, exhibits both acute and long-lasting therapeutic effects on social deficits in schizophrenia mouse models with SYT11 deficiency

    • This suggests that D2R-targeting strategies could provide comprehensive and long-term recovery for schizophrenia-associated social withdrawal

  • SYT11 level regulation in Parkinson's disease:

    • Since SYT11 accumulation mediates parkin-linked neurotoxicity, reducing SYT11 levels represents a potential therapeutic strategy

    • Targeted knockdown of SYT11 in the substantia nigra pars compacta or knockout restricted to dopaminergic neurons reverses PD-like neurotoxicity in parkin-deficient models

    • Development of small molecules that enhance SYT11 degradation or inhibit its negative effects on endocytosis could provide novel therapeutic approaches

  • Domain-specific interventions:

    • The identification of specific domains mediating SYT11 functions presents opportunities for targeted interventions

    • For example, peptides derived from the linker domain of SYT11 can compete with the interaction between SYT11 and vti1a/vti1b

    • Structure-based drug design could yield compounds that modulate specific SYT11 interactions without affecting others

  • Developmental timing considerations:

    • Given the importance of developmental timing in SYT11-related pathology, early intervention strategies targeting adolescent development periods may be particularly effective for preventing schizophrenia-related outcomes

What are the most promising biomarker applications of SYT11 in neuropsychiatric disorders?

Several biomarker applications of SYT11 show promise for neuropsychiatric disorders:

  • Expression level biomarkers:

    • SYT11 expression is reduced in individuals with schizophrenia but restored following treatment with antipsychotics

    • This suggests SYT11 levels could serve as both diagnostic and treatment response biomarkers

    • Peripheral measurements (blood cells, exosomes) could potentially reflect CNS changes

  • Genetic variation biomarkers:

    • SYT11 is listed in genetic databases as potentially associated with disease

    • Specific variants in the SYT11 gene might serve as risk predictors or treatment stratification markers

    • Comprehensive genetic screening could identify clinically relevant SYT11 variants

  • Functional biomarkers:

    • Dopamine transmission alterations resulting from SYT11 dysfunction could be measured through neuroimaging

    • PET imaging of dopamine receptors or transporters might reflect SYT11-related pathophysiology

    • fMRI measures of prefrontal cortex function could indicate downstream effects of SYT11 alterations

  • Interaction-based biomarkers:

    • Measurements of SYT11-parkin interactions or downstream effects

    • Levels of vti1a/SYT11 complexes could reflect disease state or progression

What key research questions about SYT11 remain unanswered?

Despite significant advances, several critical questions about SYT11 function remain unresolved:

  • Structural biology questions:

    • Full structural characterization of SYT11 alone and in complex with its binding partners is lacking

    • How do SYT11's non-Ca²⁺-binding C2 domains differ structurally from calcium-binding synaptotagmins?

    • What structural changes occur upon binding to partners like vti1a or parkin?

  • Developmental regulation questions:

    • What mechanisms control SYT11 expression during critical developmental periods?

    • Why does SYT11 deficiency in early adolescence, but not adulthood, lead to persistent deficits?

    • How does SYT11 contribute to normal neurodevelopmental processes?

  • Cell-type specific function questions:

    • How does SYT11 function differently across neuronal subtypes (dopaminergic, glutamatergic, GABAergic)?

    • What is the full extent of SYT11's role in microglia and other non-neuronal cells?

    • Are there region-specific functions of SYT11 in different brain areas?

  • Therapeutic translation questions:

    • Can SYT11-based interventions be effectively delivered to the CNS?

    • What are the potential off-target effects of modulating SYT11 levels or function?

    • Could SYT11-targeted approaches be combined with existing treatments for enhanced efficacy?

What control conditions are essential when manipulating SYT11 expression in experimental models?

When designing experiments involving SYT11 manipulation, several critical control conditions should be implemented:

  • Anatomical controls:

    • Unilateral manipulations with contralateral comparisons provide powerful within-subject controls

    • For example, unilateral overexpression of SYT11 in the SNpc allows comparison of dopamine release between ipsilateral and contralateral striatum

    • Adjacent brain regions should be examined to confirm spatial specificity of effects

  • Molecular specificity controls:

    • Domain-mutated or truncated SYT11 variants should be included

    • Studies have shown that C2B-truncated SYT11 fails to induce the neurotoxic effects seen with full-length SYT11

    • Rescue experiments with wild-type SYT11 following knockdown confirm specificity

  • Temporal controls:

    • Given the critical importance of developmental timing, age-matched controls are essential

    • Inducible systems should include vehicle-treated controls

    • Longitudinal assessments are valuable to distinguish between temporary and persistent effects

  • Pathway validation controls:

    • Manipulation of downstream or interacting partners

    • For example, vti1a knockdown reverses SYT11 knockout phenotypes, confirming pathway specificity

    • Pharmacological interventions targeting the same pathway should phenocopy genetic manipulations

How can researchers integrate multimodal approaches to comprehensively characterize SYT11 function?

A comprehensive characterization of SYT11 function requires integration of multiple experimental modalities:

  • Molecular and cellular approaches:

    • Protein-protein interaction studies (co-IP, GST pull-down, proximity labeling)

    • Live-cell imaging of vesicle dynamics and protein trafficking

    • Electron microscopy for ultrastructural analysis

    • Single-cell transcriptomics to identify cell type-specific effects

  • Functional assessments:

    • Electrophysiological recordings (patch-clamp, field potentials)

    • Electrochemical measurements of neurotransmitter release (amperometry with CFEs)

    • Calcium imaging to assess presynaptic function

    • Optogenetic manipulation combined with SYT11 modulation

  • Behavioral analysis:

    • Motor function assessment (especially for Parkinson's disease models)

    • Social interaction paradigms (for schizophrenia-related research)

    • Cognitive testing (learning, memory, executive function)

    • Longitudinal behavioral phenotyping to capture progressive changes

  • Translational integration:

    • Parallel studies in multiple model systems (cell culture, rodents, human samples)

    • Correlative analysis of findings across different levels of biological organization

    • Computational modeling to integrate diverse datasets and predict system-level effects

What quantitative methodologies best capture SYT11's effects on neural circuit function?

Several quantitative approaches can effectively measure SYT11's impact on neural circuits:

  • Electrochemical quantification:

    • Amperometric recordings with CFEs in striatal slices can precisely quantify dopamine release in response to local electrical stimulation

    • This technique has successfully demonstrated that unilateral SYT11 overexpression decreases ipsilateral dopamine release compared to contralateral controls

    • Key parameters include peak amplitude (~400 pA), corresponding to approximately 2.4 μM dopamine concentration

  • Electrophysiological analysis:

    • Miniature postsynaptic current analysis can quantify changes in spontaneous release

    • Parameters include event frequency and amplitude, with SYT11 specifically affecting frequency but not amplitude

    • Paired-pulse ratio measurements can assess presynaptic release probability

    • Long-term potentiation/depression protocols can evaluate synaptic plasticity

  • Network activity measurements:

    • Local field potential recordings to assess synchronized activity

    • Multi-electrode array recordings to capture circuit-level effects

    • Calcium imaging of neuronal populations to measure coordinated activity patterns

  • Computational modeling:

    • Bayesian modeling approaches to integrate diverse measurements

    • Parameter estimation techniques to quantify SYT11's contribution to circuit function

    • Network models incorporating SYT11-mediated changes in release properties

Each of these methodologies offers unique advantages in capturing different aspects of SYT11's effects on neural circuit function, from molecular interactions to systems-level outcomes.

Product Science Overview

Introduction

Synaptotagmin XI (SYT11) is a member of the synaptotagmin family, which consists of membrane-trafficking proteins characterized by an N-terminal transmembrane region, a variable linker, and two C-terminal C2 domains (C2A and C2B) . Synaptotagmins are known for their role in calcium-dependent regulation of membrane trafficking, particularly in synaptic transmission .

Structure and Function

Synaptotagmin XI, like other synaptotagmins, is involved in the regulation of neurotransmitter release. It functions as a calcium sensor, binding to calcium ions and facilitating the fusion of synaptic vesicles with the presynaptic membrane . This process is crucial for the release of neurotransmitters into the synaptic cleft, enabling communication between neurons.

Expression and Localization

SYT11 is predominantly expressed in brain tissue, where it plays a pivotal role in neuronal vesicular trafficking and exocytosis . It is distributed throughout the neuronal cell body, axons, and dendrites, mediating vesicular trafficking and ensuring efficient neurotransmitter release .

Clinical Significance

Recent studies have highlighted the clinical significance of SYT11 in various cancer types. Abnormal expression of SYT11 has been observed in different tumor types, and its expression levels have been correlated with patient prognosis . Additionally, SYT11 has been implicated in immune infiltration and various molecular pathways, including clathrin-mediated endocytosis and Rho GTPase signaling .

Research and Therapeutic Potential

Research on SYT11 has revealed its potential as a therapeutic target. Genome-associated studies and experimental models have shown that SYT11 dysfunction is associated with neurological disorders such as Parkinson’s disease and schizophrenia . Understanding the molecular mechanisms underlying SYT11’s role in these diseases could pave the way for the development of novel therapeutic strategies.

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.