Enhanced LTP in Knockout Models: S100B-deficient mice exhibit strengthened long-term potentiation (LTP) in hippocampal CA1 regions and superior spatial memory in Morris water maze tests .
Extracellular Modulation: Perfusion of recombinant S100B (1 µg/mL) normalizes LTP levels in mutant hippocampal slices, confirming its extracellular regulatory role .
Blood-Brain Barrier (BBB) Integrity: S100B knockout mice develop chronic BBB permeability defects, highlighting its role in maintaining barrier function .
EAE Model Relevance: In experimental autoimmune encephalomyelitis (EAE), S100B deletion reduces demyelinating lesions and improves motor outcomes, suggesting therapeutic potential in multiple sclerosis .
Astrocyte and Microglia Regulation: Recombinant S100B modulates glioma cell migration and microglial activation, as shown in GL261 murine models .
Calcium Signaling: Acts as a calcium sensor, influencing kinase pathways (e.g., STK38) and apoptosis via ERK1/2 and p53 signaling .
Fear Conditioning: S100B-null mice display heightened contextual fear memory, with freezing rates 20–30% higher than wild-type counterparts .
Epileptogenesis: Elevated S100B levels correlate with reduced seizure severity, indicating neuroprotective effects .
Concentration and Formulation: Supplied at 1 mg/mL in 20 mM Tris-HCl buffer (pH 8.0) with 1 mM DTT and 10% glycerol .
Endotoxin Levels: <1 EU per µg, ensuring suitability for cell culture and in vivo studies .
Functional Validation: Confirmed via SDS-PAGE, MALDI-TOF, and activity assays in synaptic plasticity models .
S100B transgenic (S100Btg) mice with multiple copies of the murine S100B gene are widely used for studying long-term increased S100B expression. The most documented model contains 12 copies of the murine S100B gene, resulting in nanomolar S100B concentrations in brain tissue, cerebrospinal fluid (CSF), and serum . These levels remain below the toxic micromolar concentrations typically used in cell culture experiments.
In contrast, S100B knockout (S100B KO) mice have been developed to study the effects of S100B deletion. These models show almost non-existent tissue S100B expression compared to wild-type counterparts .
For studying specific mechanisms, researchers should consider:
S100Btg mice: Optimal for investigating neurotrophic effects at nanomolar concentrations
S100B KO mice: Valuable for understanding loss-of-function consequences, particularly in neuroinflammatory conditions
His-tagged S100B recombinant proteins typically contain a hexahistidine tag (HHHHHH) fused to the S100B protein sequence. This modification facilitates protein purification through affinity chromatography but may influence protein behavior in some experimental settings.
Key differences include:
Molecular weight: His-tagged mouse S100B recombinant proteins have a predicted molecular weight of approximately 12.8 kDa (112 amino acids), compared to native S100B (approximately 10.5 kDa)
Formulation: Recombinant His-tagged S100B is typically supplied in buffer solutions containing stabilizers (e.g., 20mM Tris-HCl buffer pH 8.0 with 1mM DTT, 10% glycerol)
Purity: Commercial preparations are typically >85% pure as assessed by SDS-PAGE
For physiologically relevant experiments, researchers should consider whether the His-tag might interfere with binding to target receptors or downstream signaling pathways.
To ensure experimental rigor with S100B mouse models, researchers should validate:
S100B expression levels: Confirm elevated S100B in transgenic models or absent expression in knockout models using techniques like western blotting or qPCR
Age-dependent effects: S100B effects vary significantly across juvenile (28 days), adult (3 months), and senile (one year) mice
Sex-specific differences: Male and female S100Btg mice show differential effects on ApoE and BDNF expression
Regional expression patterns: Assess S100B levels in relevant brain regions (hippocampus, frontal cortex)
Baseline cognitive and behavioral phenotypes: Document any baseline differences in neurological function
Data from transgenic models indicate approximately 50% increased proliferation in juvenile S100Btg mice and 200% increased proliferation in adult S100Btg mice compared to wild-type controls .
Quantifying hippocampal neurogenesis in S100B mouse models requires careful methodological consideration:
Method comparison:
Both BrdU incorporation and Ki67 immunoreactivity provide reliable measures of progenitor cell proliferation. Studies have demonstrated a significant correlation between these methods (r = 0.608, p < 0.001) .
Recommended protocol:
For BrdU labeling: Administer 50 mg/kg BrdU intraperitoneally before tissue collection
For Ki67 immunohistochemistry: Use standard immunohistochemical procedures on fixed tissue sections
Quantify proliferating cells specifically in:
Subgranular zone (SGZ) - The primary germinative area of the dentate gyrus
Granular cell layer (GCL) - The destination area for migrating progenitor cells
Compare multiple age groups to account for age-dependent decline in neurogenesis
Ki67 immunohistochemistry may be preferable as it avoids the experimental burden of BrdU injections while providing comparable results, aligning with EU Directive 2010/63/EU on reducing animal experimentation burden .
To comprehensively evaluate S100B's role in neurogenesis, researchers should assess the following markers:
Proliferation markers:
Ki67: Endogenous marker expressed during all active phases of the cell cycle
BrdU: Exogenous thymidine analog that incorporates into newly synthesized DNA
Associated signaling proteins:
GFAP (Glial Fibrillary Acidic Protein): Marker for astrocytes and neural stem cells
ApoE (Apolipoprotein E): Associated with lipid metabolism and neuronal repair
BDNF (Brain-Derived Neurotrophic Factor): Growth factor that promotes neuronal survival
RAGE (Receptor for Advanced Glycation End Products): Principal receptor for S100B
Neuroinflammatory markers:
Iba1: General microglia/macrophage marker
iNOS: Marker for pro-inflammatory microglia/macrophages
CX3CR1 (Fractalkine receptor): Marker for homeostatic/phagocytic microglia
Studies have shown that long-term increased S100B levels correlate with decreased hippocampal GFAP, suggesting reduced astrogliosis rather than increased reactive astrocytes .
For His-tagged S100B protein analysis:
Purity assessment:
SDS-PAGE: Verify a single band at approximately 12.8 kDa (for His-tagged mouse S100B)
MALDI-TOF: Confirm predicted molecular weight of 12.8 kDa for the 112 amino acid sequence
Endotoxin testing: Ensure levels are <1 EU per 1μg of protein (using LAL method)
Activity testing:
Binding assays: Confirm interaction with known S100B receptors (e.g., RAGE)
Functional assays: Test effects on cell proliferation in neural progenitor cultures
Downstream signaling: Assess activation of known S100B-dependent pathways
Storage and handling:
Short-term (1 week): Store at 2-8°C
Long-term: Aliquot and store at -20°C to -80°C
S100B demonstrates complex concentration-dependent and age-dependent effects on hippocampal neurogenesis:
Concentration effects:
Nanomolar (nM) concentrations: Neurotrophic, promoting progenitor cell proliferation
Micromolar (μM) concentrations: Typically toxic in cell culture models
Age-dependent effects (Ki67+ cells in SGZ):
Age Group | Wild-type | S100Btg | Percent Increase |
---|---|---|---|
Juvenile (28 days) | Variable | +50% | 50% |
Adult (3 months) | Lower than juvenile | +200% | 200% |
One-year-old | Nearly undetectable | 0.24±0.07 cells | Significant |
S100B-induced progenitor cell proliferation in the SGZ increases with age relative to the declining baseline in wild-type mice. Additionally, migration of proliferating cells to the GCL increases with age in S100Btg mice .
The mechanism appears to involve direct S100B effects rather than indirect actions through BDNF, as hippocampal BDNF levels remain unchanged in S100Btg mice despite enhanced neurogenesis .
Research reveals important sex-specific differences in S100B function:
Male-specific effects:
Reduced ApoE levels in both hippocampus and frontal cortex of male S100Btg mice
Female-specific effects:
Reduced BDNF in frontal cortex of female S100Btg mice
Sex-independent effects:
Enhanced hippocampal progenitor cell proliferation in both sexes
Methodological recommendations:
Always include both sexes in experimental designs
Analyze data separately by sex before pooling
Consider potential hormonal influences on S100B signaling
When studying ApoE or BDNF interactions with S100B, sex stratification is crucial
For translational studies, sex differences may have implications for therapeutic approaches
These findings highlight the complex sex-dependent cross-talk between S100B and other signaling molecules relevant to neurogenesis and neuroinflammation.
S100B plays a complex role in neuroinflammatory conditions as demonstrated in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis:
S100B deletion effects in EAE:
Delayed disease onset
Reduced paralysis
Accelerated recovery, particularly in chronic stages
Neuroinflammatory marker changes:
Reduced GFAP expression (astrocyte marker) in S100B KO EAE mice
Reduced Iba1 expression (microglia/macrophage marker) in S100B KO EAE mice
Decreased iNOS (pro-inflammatory marker) expression
Altered CX3CR1 (homeostatic/phagocytic microglia marker) expression
S100B functions as a damage-associated molecular pattern (DAMP) or alarmin, released upon tissue injury and contributing to neuroinflammation. Research suggests therapeutic potential in blocking S100B in neuroinflammatory conditions, as demonstrated by pentamidine treatment in EAE models .
The dual role of S100B (neurotrophic at nanomolar levels, potentially harmful in inflammatory conditions) highlights the importance of context-specific research approaches.
Research on S100B inhibition has revealed promising therapeutic potential:
In multiple sclerosis models:
S100B knockout mice show ameliorated clinical symptoms in EAE
Pharmacological inhibition with pentamidine changed disease course and improved outcome
Pentamidine treatment prevented lesion formation and oligodendrocyte impairment
S100B inhibition decreased astrocytic reactivity and promoted non-pro-inflammatory microglia/macrophages
Immunomodulatory effects:
Enhanced CNS infiltration by regulatory T (Treg) cells
Reduced T helper (Th)1 and Th17 cell infiltration
These findings suggest repurposing pentamidine or developing other S100B inhibitors as potential therapies for multiple sclerosis and potentially other neuroinflammatory conditions.
The seemingly contradictory findings from S100B transgenic and knockout models can be reconciled to understand therapeutic potential:
Integrative model:
Context-dependent effects: S100B acts as neurotrophic at nanomolar levels in non-inflammatory contexts, while functioning as a DAMP in inflammatory conditions
Disease-specific considerations: In traumatic brain injury or developmental contexts, promoting S100B may enhance neurogenesis; in neuroinflammatory conditions, inhibiting S100B may be beneficial
Concentration-dependent paradigm: Therapeutic approaches should consider whether local S100B concentrations are in the neurotrophic (nM) or potentially harmful (μM) range
Research recommendations:
Combine transgenic overexpression studies with controlled inhibition experiments
Monitor both S100B levels and inflammatory markers
Test therapeutic hypotheses in disease-specific models
Consider sex as a biological variable, given sex-specific effects
Evaluate age-dependent effects, as S100B function changes across the lifespan
This integrated approach can help reconcile the dual roles of S100B and guide development of context-appropriate therapeutic strategies.
To evaluate His-tagged S100B as a potential therapeutic agent:
In vitro assessments:
Dose-response studies (nM vs. μM concentrations) on neural progenitor cultures
Receptor binding studies (particularly RAGE) with purified His-tagged protein
Verification that His-tag doesn't interfere with biological activity
Stability testing under physiological conditions
In vivo approaches:
Intrathecal or intraperitoneal administration protocols
Blood-brain barrier penetration studies
Pharmacokinetic assessment of His-tagged S100B
Functional outcome measures following administration:
Hippocampal-dependent cognitive tasks
Histological assessment of neurogenesis (Ki67/BrdU)
Evaluation of associated markers (GFAP, ApoE, BDNF)
Prior research has demonstrated that "acute intrathecal and intraperitoneal treatment with S100B in wild-type rodents promoted hippocampal neurogenesis and cognitive function," suggesting potential therapeutic applications in contexts requiring enhanced neurogenesis.
S100 Calcium Binding Protein B (S100B) is a member of the S100 protein family, which is characterized by the presence of two EF-hand calcium-binding motifs. These proteins are localized in the cytoplasm and/or nucleus of a wide range of cells and are involved in the regulation of various cellular processes such as cell cycle progression and differentiation .
The S100B gene is located on chromosome 21q22.3 in humans and has orthologs in various species, including mice. The protein encoded by this gene is known for its role in the central nervous system (CNS), particularly in astrocytes, which are a type of glial cell . The recombinant form of S100B, tagged with a His (histidine) tag, is often used in research to facilitate purification and detection.
S100B has several critical functions:
S100B is secreted by astrocytes and can spill from injured cells into the extracellular space or bloodstream. Elevated levels of S100B in the serum are indicative of acute brain damage and have been used as a biomarker for blood-brain barrier (BBB) permeability and CNS injury. Elevated S100B levels are associated with neuropathological conditions such as traumatic head injury and neurodegenerative diseases .
The recombinant form of S100B, tagged with a His tag, is produced in a laboratory setting to facilitate research. The His tag allows for easy purification and detection of the protein using affinity chromatography techniques. This recombinant protein is used in various studies to understand the functions and mechanisms of S100B in both normal and disease states.