S100G Rat

S100 Calcium Binding Protein G Rat Recombinant
Shipped with Ice Packs
In Stock

Description

Calcium Homeostasis

S100G facilitates intestinal calcium absorption by buffering cytoplasmic Ca²⁺ and enhancing ATP-dependent transport in duodenal membranes . In Sprague Dawley rats, serum calcium levels average 12.3 ± 0.4 mg/dL (males) and 11.4 ± 3.5 mg/dL (females) , with S100G contributing to maintaining these levels.

Reproductive System

S100G mediates estrogen (E2)-induced genomic effects, accelerating egg transport in the rat oviduct. It is upregulated in the pituitary gland by estrogen, linking it to reproductive hormone regulation .

Nervous System

S100G is expressed in distinct neuronal subpopulations, including brainstem and cerebellar neurons. Its immunoreactivity varies with functional states, suggesting roles in neuronal Ca²⁺ signaling and stress responses .

Neurodevelopmental Disorders

Rat studies implicate S100G in autism spectrum disorder and syndromic X-linked intellectual disability (Lubs type), potentially via dysregulated calcium signaling .

Liver Disease

While S100A4/A9 dominate liver pathology research, S100G’s involvement in hepatic Ca²⁺ metabolism warrants further study .

Table 1: Functional Interactions of S100G in Rats

TargetInteractionFunctional OutcomeSource
ATP-dependent Ca²⁺ pumpsEnhances activityImproved duodenal Ca²⁺ transport
Estrogen receptorsE2-induced upregulationAccelerated oviductal egg transport
Reactive oxygen speciesScavenging via Ca²⁺ bufferingReduced oxidative stress in neurons

Table 2: Expression Levels in Rat Tissues

TissueExpression LevelRegulatory Factor
DuodenumHighVitamin D, dietary Ca²⁺
UterusModerateEstrogen
Pituitary glandModerateHormonal stimuli
Cerebral neuronsVariableFunctional state, stress

Implications for Biomedical Research

S100G’s role as a calcium buffer makes it a potential therapeutic target for disorders involving Ca²⁺ dysregulation, such as neurodegenerative diseases or metabolic bone disorders. Its estrogen-responsive expression also highlights its relevance in reproductive health studies . Future research should explore its interactions with other S100 proteins and extracellular receptors in rat models .

Product Specs

Introduction
Calretinin, a calcium-binding protein of the troponin C superfamily, is characterized by EF-hand domains. In the developing cerebellum, immunohistochemical detection of calretinin reveals weak staining from week 21 of gestation, primarily in Purkinje, basket cells, and neurons of the dentate nucleus. This staining intensifies with cerebellar maturation. Calretinin expression is also observed in tumors, particularly mesotheliomas and certain pulmonary adenocarcinomas. In the context of recombinant protein production, S100G, a calcium-binding protein found in rat, serves as a comparable protein.
Description
S100G, also known as Calbindin-D9k or CaBP9K, is expressed with a -6xHis tag and purified by proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered White lyophilized (freeze-dried) powder.
Formulation
The S100G protein was lyophilized from a concentrated solution (1mg/ml) containing 100mM Phosphate buffer, pH 7.3.
Solubility
It is recommended to reconstitute the lyophilized S100G in sterile 18MΩ-cm H2O not less than 100µg/ml, which can then be further diluted to other aqueous solutions.
Stability
Lyophilized S100G although stable at room temperature for 3 weeks, should be stored desiccated below -18°C. Upon reconstitution S100G should be stored at 4°C between 2-7 days and for future use below -18°C. For long term storage it is recommended to add a carrier protein (0.1% HSA or BSA).
Please prevent freeze-thaw cycles.
Purity
Greater than 90% as determined by SDS-PAGE.
Synonyms
Protein S100-G, S100 calcium-binding protein G, Vitamin D-dependent calcium-binding protein intestinal, CABP, Calbindin-D9k, 9 kDa CaBP, Cholecalcin, S100g, Calb3, S100d, Cbpi, MGC72928, Rncalbd9.
Source
Escherichia Coli.

Q&A

What is S100G and what is its significance in rat research models?

S100G, also known as CalbindinD-9K, is a calcium-binding protein belonging to the S100 family. In rat models, it serves as an important calcium buffer, playing crucial roles in calcium homeostasis across various tissues. The significance of S100G in rat research stems from its involvement in critical physiological processes including calcium absorption, transportation, and buffering. S100G is particularly valuable in translational research due to its conserved structure and function across mammalian species, making rat models relevant for understanding human pathophysiology related to calcium regulation disorders .

How is S100G expression distributed across rat tissues?

S100G expression in rats demonstrates a tissue-specific distribution pattern that varies with developmental stage and physiological conditions. While early studies suggested that S100 proteins were brain-specific, current research confirms their presence in multiple tissues. S100G mRNA has been detected in numerous rat tissues beyond the central nervous system. In the brain, S100 proteins (including S100G) are synthesized in all astrocytes and in distinct subpopulations of neurons with characteristic topographical distribution .

Within the central nervous system, there are two distinct classes of S100-positive neuron populations:

  • "Persistently S100-positive" neurons: Large, strongly immunoreactive neurons primarily distributed in the nuclei of the lower brainstem and cerebellum

  • "Variably S100-positive" neurons: Predominantly found in the forebrain of rats older than 90 days, particularly abundant in limbic regions, with moderate immunoreactivity that shows high interindividual variation

In addition to neural tissues, S100G is expressed in intestinal tissues and pancreatic acini, where its expression can be induced under pathological conditions such as pancreatitis .

What are the fundamental methodological approaches for studying S100G in rat models?

When studying S100G in rat models, researchers should consider multiple complementary methodological approaches:

  • Gene expression analysis:

    • Real-time quantitative PCR (qPCR) using validated primers specific for the S100g gene

    • Microarray analysis for high-throughput gene expression profiling

    • In situ hybridization to visualize spatial expression patterns

  • Protein detection:

    • Immunohistochemistry and immunofluorescence for tissue localization

    • Western blotting for protein quantification

    • ELISA for sensitive protein measurement

  • Functional analysis:

    • Calcium imaging techniques to assess calcium buffering

    • Knockout or knockdown models to evaluate physiological roles

    • Overexpression studies to assess effects of elevated S100G levels

A comprehensive approach typically involves correlating gene expression with protein levels across multiple tissues and under various physiological and pathological conditions. It's important to note that direct correlations between protein and mRNA levels are not always observed, suggesting that different mechanisms regulate S100G expression in various tissues .

How should researchers design experiments to investigate the role of S100G in calcium regulation during pancreatitis?

When investigating S100G's role in calcium regulation during pancreatitis, a well-designed experimental approach should include:

Experimental model selection:

  • Cerulein-induced pancreatitis is the recommended model as it reliably induces S100G expression in pancreatic acini

  • AR42J cells (rat pancreatic acinar cell line) can be used for in vitro studies to complement in vivo experiments

Experimental design structure:

  • Baseline characterization: Measure baseline S100G expression in healthy pancreatic tissue

  • Disease progression monitoring: Track temporal changes in S100G expression at multiple timepoints (early, peak, and resolution phases)

  • Intervention studies: Test the effects of calcium channel blockers or calcium chelators on S100G expression and pancreatitis severity

  • Mechanistic studies: Investigate the interaction between S100G and other calcium regulatory proteins

Key measurements:

  • S100G gene and protein expression levels

  • Intracellular calcium concentration

  • Markers of pancreatic injury (amylase, lipase)

  • Inflammatory mediators

  • Acinar cell death assessment

Controls and validation:

  • Include both positive controls (animals with confirmed pancreatitis) and negative controls (sham-treated animals)

  • Validate S100G expression changes using multiple techniques (qPCR, Western blot, immunohistochemistry)

  • Consider using S100G knockout or transgenic rat models if available

What are the optimal methodologies for differentiating between S100G and other S100 family proteins in rat tissues?

Differentiating between S100G and other S100 family proteins requires careful methodological consideration due to their structural similarities. The following approaches are recommended:

Molecular techniques:

  • Primer and probe selection for qPCR: Design primers that target unique regions of S100G mRNA not shared with other S100 family members. Validate specificity using positive and negative controls. Commercial assays like Bio-Rad's PrimePCR assays for S100g provide validated primers optimized for specificity .

  • Antibody selection for protein detection:

    • Use monoclonal antibodies specifically raised against S100G

    • Perform pre-absorption controls with recombinant S100G to confirm specificity

    • Include appropriate controls (tissues known to express or lack S100G)

  • Protein separation techniques:

    • 2D gel electrophoresis can separate S100 proteins based on both molecular weight and isoelectric point

    • Mass spectrometry for definitive identification of S100G

Validation approaches:

  • Cross-validate results using multiple antibodies targeting different epitopes

  • Compare expression patterns with published distribution maps of S100 proteins

  • Use S100G knockout tissue as a negative control when available

Common pitfalls to avoid:

  • Commercial antibodies may cross-react with other S100 family members; validate specificity before use

  • Low expression levels may require more sensitive detection methods

  • RNA probes and PCR primers should be designed to account for potential splice variants

How can researchers effectively study S100G function using gene modification approaches in rats?

Studying S100G function through gene modification requires careful planning and methodological precision:

Gene modification strategies:

  • Knockout approaches:

    • CRISPR/Cas9 system for targeted S100G gene deletion

    • Conditional knockout models using Cre-loxP systems for tissue-specific or inducible deletion

    • Evaluation of compensatory mechanisms, as other calcium transporter genes may be induced when S100G is deleted

  • Knockdown approaches:

    • siRNA or shRNA delivery for transient or stable S100G reduction

    • Antisense oligonucleotides for targeted mRNA degradation

  • Overexpression approaches:

    • Viral vectors (adenovirus, lentivirus) for S100G overexpression

    • Transgenic rat development with tissue-specific promoters

Experimental considerations:

  • Phenotype assessment: Careful evaluation of phenotype is essential, as studies have shown that S100G knockout mice are indistinguishable from wild-type mice in phenotype and serum calcium level due to compensatory mechanisms

  • Tissue-specific effects: Focus on tissue-specific consequences of gene modification rather than just systemic calcium levels

  • Functional redundancy: Address potential functional redundancy with other calcium-binding proteins

  • Timing: Consider developmental timing of gene modification, as effects may differ between embryonic and adult stages

Validation approaches:

  • Confirm genetic modification at both DNA and RNA levels

  • Verify protein expression changes using Western blot and immunohistochemistry

  • Evaluate calcium handling using calcium imaging techniques

  • Assess physiological impact through functional tests appropriate to the tissue under study

How should researchers address contradictory findings regarding S100G expression patterns in rat tissues?

Contradictory findings regarding S100G expression are common in the literature and require systematic approaches for resolution:

Sources of potential contradictions:

  • Methodological variations:

    • Different antibody specificities and sensitivities

    • Varied detection thresholds of PCR techniques

    • Differences in tissue preparation and fixation

  • Biological variables:

    • Age and developmental stage differences (expression patterns change with maturation)

    • Strain differences between rat lines

    • Sex-dependent expression patterns

    • Functional state of the tissue at sample collection

Systematic approach to resolve contradictions:

  • Comparative methodology analysis:

    • Use multiple detection methods in parallel (e.g., qPCR, Western blot, immunohistochemistry)

    • Compare results obtained with different antibodies or primer sets

    • Establish detection thresholds and quantification standards

  • Standardized reporting:

    • Document detailed experimental conditions including rat strain, age, sex, and tissue processing methods

    • Report specific reagents including antibody clones, dilutions, and incubation conditions

    • Include both positive and negative control tissues

  • Contextual interpretation:

    • Consider functional state of the tissue (e.g., S100G expression in neurons varies with functional activity)

    • Account for interindividual variation, which is particularly high in "variably S100-positive" neurons

    • Compare findings across multiple experimental models and conditions

When evaluating contradictory literature, researchers should construct a comparative table documenting methodological details, rat characteristics, and specific findings across studies to identify patterns that may explain discrepancies .

What statistical approaches are most appropriate for analyzing S100G expression changes in different experimental conditions?

The statistical analysis of S100G expression data requires careful consideration of data characteristics and experimental design:

Recommended statistical approaches:

Data normalization considerations:

  • For qPCR data: Identify stable reference genes specific to the experimental conditions

  • For Western blot: Normalize to loading controls (β-actin, GAPDH) with verification of linear range

  • For immunohistochemistry: Use appropriate internal controls and standardized quantification methods

Sample size determination:

  • Conduct power analysis based on pilot data or literature-reported effect sizes

  • Consider biological variability, which is particularly high for S100G expression in certain tissues

  • Include sufficient biological replicates (different animals) and technical replicates

Reporting standards:

  • Report exact p-values rather than p-value ranges

  • Include measures of effect size and confidence intervals

  • Provide complete descriptive statistics (mean, median, standard deviation, interquartile range)

How can researchers differentiate between direct S100G effects and indirect consequences in experimental models?

Distinguishing direct S100G effects from indirect consequences requires carefully designed experimental approaches:

Methodological strategies:

  • Temporal resolution studies:

    • Track the sequence of molecular events following S100G manipulation

    • Use time-course experiments with frequent sampling to establish cause-effect relationships

    • Employ kinetic modeling to establish temporal dependencies

  • Spatial correlation approaches:

    • Co-localization studies to identify spatial relationships between S100G and potential interacting partners

    • Single-cell analysis to correlate S100G levels with cellular phenotypes

    • Tissue-specific manipulation of S100G expression

  • Molecular interaction studies:

    • Co-immunoprecipitation to identify direct protein interactions

    • Proximity ligation assays to detect protein-protein interactions in situ

    • FRET/BRET analysis for real-time interaction monitoring

  • Dose-dependency analysis:

    • Use graded levels of S100G manipulation to establish dose-response relationships

    • Correlate S100G levels with functional outcomes

    • Compare partial versus complete knockdown effects

Experimental controls:

  • Include rescue experiments where S100G is re-introduced after knockdown

  • Use structurally similar but functionally distinct S100 family members as controls

  • Employ pharmacological inhibitors of downstream pathways to block indirect effects

Data interpretation framework:

  • Direct effects typically occur rapidly and without requiring intermediate steps

  • Indirect effects often show delayed kinetics and may be blocked by inhibiting intermediate pathways

  • Consider constructing pathway maps to visualize potential direct and indirect relationships

How does S100G expression in rat models of pancreatitis correlate with disease progression and severity?

S100G expression demonstrates specific patterns in relation to pancreatitis progression and severity in rat models:

Expression patterns during disease progression:

In cerulein-induced pancreatitis, S100G expression is dynamically regulated:

  • Baseline: Low expression in healthy pancreatic tissue

  • Early phase (0-12 hours): Rapid induction of S100G expression in acinar cells

  • Peak phase (24-48 hours): Maximal expression correlating with peak inflammation

  • Resolution phase: Gradual return to baseline levels as tissue healing occurs

Correlation with severity markers:

Disease ParameterCorrelation with S100GStatistical Significance
Acinar cell injuryPositive correlationp < 0.05
EdemaPositive correlationp < 0.05
Inflammatory infiltrationPositive correlationp < 0.01
Serum amylase/lipasePositive correlationp < 0.01
Calcium dysregulationStrong positive correlationp < 0.001

Functional implications:

  • Alteration of normal calcium signaling pathways

  • Modification of inflammatory response regulation

  • Influence on acinar cell survival and apoptosis mechanisms

Clinical relevance:

The expression pattern and correlation with disease severity suggest that S100G could serve as a potential biomarker for pancreatitis progression and a target for therapeutic intervention. When evaluating therapeutic approaches, monitoring S100G expression may provide insights into intervention efficacy .

What are the most effective approaches for investigating S100G interactions with other calcium regulatory proteins in rat models?

Investigating S100G interactions with other calcium regulatory proteins requires a multi-faceted approach:

Protein interaction mapping strategies:

  • In vitro interaction studies:

    • Pull-down assays with purified recombinant proteins

    • Surface plasmon resonance to determine binding kinetics

    • Isothermal titration calorimetry for thermodynamic characterization

  • Intracellular interaction studies:

    • Co-immunoprecipitation from tissue or cell lysates

    • Proximity ligation assay for visualizing interactions in situ

    • FRET/BRET analysis for live cell interaction monitoring

    • BiFC (Bimolecular Fluorescence Complementation) for direct visualization of protein complexes

  • Functional interaction assessment:

    • Calcium imaging in the presence/absence of S100G and partner proteins

    • Competitive binding assays to identify shared binding partners

    • Electrophysiological measurements to assess functional consequences

Key protein interactions to investigate:

  • Annexin family proteins: Particularly Annexin A10, which shows coordinated expression with S100G in pancreatitis

  • Other calcium binding proteins: Including calmodulin, troponin C, and other S100 family members

  • Calcium channels and transporters: Such as TRPV6, which functions in calcium absorption pathways together with S100G

  • Signaling proteins: Kinases and phosphatases that may regulate S100G function

Experimental design considerations:

  • Use appropriate controls including calcium chelators (EGTA, BAPTA) and competitors

  • Consider both calcium-dependent and calcium-independent interactions

  • Evaluate interactions under physiological and pathological conditions

  • Employ both overexpression and knockdown approaches to validate interactions

How can researchers leverage transcriptomic approaches to understand the regulatory networks controlling S100G expression in rats?

Transcriptomic approaches offer powerful tools for elucidating the regulatory networks controlling S100G expression:

Recommended transcriptomic methodologies:

  • RNA-Seq analysis:

    • Whole transcriptome profiling to identify co-regulated genes

    • Differential expression analysis across tissues and conditions

    • Alternative splicing analysis to identify tissue-specific isoforms

  • ChIP-Seq and ATAC-Seq:

    • Identify transcription factors binding to the S100G promoter

    • Map chromatin accessibility at the S100G locus

    • Characterize enhancer elements regulating tissue-specific expression

  • Single-cell RNA-Seq:

    • Define cell-type specific expression patterns

    • Identify rare cell populations expressing S100G

    • Track transcriptional changes during differentiation or disease progression

  • Spatial transcriptomics:

    • Map S100G expression within tissue architectural context

    • Correlate expression with microenvironmental factors

Data integration approaches:

  • Network analysis:

    • Construct gene co-expression networks to identify modules containing S100G

    • Perform transcription factor enrichment analysis

    • Identify signaling pathways converging on S100G regulation

  • Multi-omics integration:

    • Correlate transcriptomic data with proteomic profiles

    • Integrate with epigenomic data (DNA methylation, histone modifications)

    • Incorporate metabolomic data to link metabolic state with S100G expression

Validation strategies:

  • Confirm key regulatory relationships using reporter assays

  • Perform targeted manipulation of identified regulators

  • Validate in multiple experimental models and conditions

Example findings from transcriptomic studies:

In rat models of rotavirus infection, gene expression analysis identified distinct patterns of gene regulation, including immunity markers and intestinal maturation genes. Similar approaches can be applied to understand S100G regulation in different physiological and pathological contexts .

What are the current challenges and future directions in S100G research using rat models?

Current challenges and future directions in S100G research with rat models encompass several key areas:

Current challenges:

  • Functional redundancy: Compensatory mechanisms activated in S100G knockout models make it difficult to isolate specific functions. For example, studies have shown that Calbindin-D9k knockout mice are indistinguishable from wild-type mice in phenotype and serum calcium level, suggesting compensation by other calcium transporter genes .

  • Tissue-specific roles: S100G appears to have distinct functions in different tissues, requiring specialized approaches for each tissue context.

  • Translational relevance: Establishing clear connections between rat model findings and human pathophysiology remains challenging.

  • Technical limitations: Current techniques may not fully capture the dynamics of calcium buffering in real-time in vivo.

Future research directions:

  • Development of conditional and inducible models:

    • Tissue-specific and temporally controlled S100G manipulation

    • Dual knockout models targeting S100G and compensatory proteins

    • Development of knock-in models with tagged S100G for tracking

  • Advanced imaging approaches:

    • Real-time calcium imaging in living animals

    • Super-resolution microscopy to visualize S100G localization and trafficking

    • Multiplexed imaging to simultaneously track multiple calcium regulatory proteins

  • Disease-focused applications:

    • Exploration of S100G's role in additional pathological conditions beyond pancreatitis

    • Development of S100G-targeted therapeutic approaches

    • Investigation of S100G as a biomarker for disease progression and treatment response

  • Multi-omics integration:

    • Comprehensive profiling of S100G-expressing cells across multiple molecular levels

    • Systems biology approaches to model calcium regulatory networks

    • Artificial intelligence-based prediction of S100G function and regulation

  • Comparative studies:

    • Cross-species comparisons to establish evolutionary conservation of S100G functions

    • Examination of S100G variants and their functional consequences

What are the optimal sample preparation techniques for reliable S100G protein detection in rat tissues?

Sample preparation is critical for reliable S100G protein detection, with specific considerations for different rat tissues:

Tissue collection and preservation:

  • Fresh tissue collection:

    • Rapid extraction and processing minimizes protein degradation

    • Flash freezing in liquid nitrogen is recommended for molecular analysis

    • Careful dissection to isolate specific regions, particularly important for brain tissue with region-specific expression patterns

  • Fixation for immunohistochemistry:

    • 4% paraformaldehyde provides good antigen preservation while maintaining tissue architecture

    • Fixation time should be optimized (typically 24-48 hours for whole organs, 4-24 hours for tissue slices)

    • Post-fixation washing is essential to remove excess fixative

  • Tissue storage considerations:

    • For protein analysis: Store at -80°C with protease inhibitors

    • For fixed tissues: Store in 30% sucrose solution at 4°C for short-term or in cryoprotectant at -20°C for long-term

Protein extraction protocols:

  • For total protein extraction:

    • RIPA buffer supplemented with protease inhibitors and phosphatase inhibitors

    • Homogenization using mechanical disruption (e.g., homogenizer, sonication)

    • Centrifugation to remove debris (12,000-15,000 g for 15-20 minutes at 4°C)

  • For subcellular fractionation:

    • Sequential extraction using buffers of increasing strength

    • Differential centrifugation to isolate cellular compartments

    • Sucrose gradient centrifugation for membrane fraction isolation

  • For preservation of protein-protein interactions:

    • Milder lysis buffers containing non-ionic detergents (e.g., NP-40, Triton X-100)

    • Chemical crosslinking before lysis for transient interactions

    • Native extraction conditions when possible

Special considerations for S100G:

  • As a calcium-binding protein, calcium chelators like EDTA or EGTA in buffers may alter S100G conformation and affect detection

  • Include both calcium-free and calcium-containing conditions when optimizing extraction

  • Consider native gel electrophoresis to preserve calcium-dependent conformations

What are the key considerations for designing qPCR experiments to quantify S100G gene expression in rat models?

Designing qPCR experiments for S100G quantification requires careful consideration of several key factors:

Primer and probe design:

  • Target sequence selection:

    • Target unique regions of S100G not present in other S100 family members

    • Avoid regions with known polymorphisms or splice variants

    • Design primers spanning exon-exon junctions to avoid genomic DNA amplification

  • Primer characteristics:

    • Optimal length: 18-22 nucleotides

    • GC content: 40-60%

    • Melting temperature: 58-62°C with minimal difference between forward and reverse primers

    • Avoid secondary structures, primer dimers, and repeat sequences

  • Validation requirements:

    • Test primer efficiency using standard curves (efficiency should be 90-110%)

    • Verify specificity through melt curve analysis and gel electrophoresis

    • Consider using validated commercial assays like Bio-Rad's PrimePCR assays for S100g

Reference gene selection:

  • Stability assessment:

    • Evaluate multiple candidate reference genes across experimental conditions

    • Use algorithms like geNorm, NormFinder, or BestKeeper to identify the most stable references

    • Common reference genes include β-actin, GAPDH, and 18S rRNA, but their stability must be verified for specific experimental conditions

  • Optimal normalization strategy:

    • Use multiple reference genes rather than a single reference

    • Calculate geometric mean of reference genes for normalization

    • Consider using global normalization approaches for large-scale studies

Experimental design considerations:

  • Sample processing:

    • Extract RNA using methods that preserve integrity (RIN > 7 recommended)

    • Include DNase treatment to remove genomic DNA contamination

    • Use consistent RNA input amounts for reverse transcription

  • Controls:

    • No-template controls to detect contamination

    • No-reverse transcriptase controls to detect genomic DNA contamination

    • Positive controls (tissues known to express S100G)

    • Inter-run calibrators for studies requiring multiple PCR runs

  • Replication strategy:

    • Include both biological replicates (different animals) and technical replicates

    • Minimum of 3 biological replicates per experimental group

    • 2-3 technical replicates per biological sample

Data analysis approach:

  • Use appropriate quantification methods (standard curve or comparative Ct method)

  • Apply correct statistical tests based on data distribution

  • Report results according to MIQE guidelines (Minimum Information for Publication of Quantitative Real-Time PCR Experiments)

How can researchers effectively compare S100G expression across different rat strains and developmental stages?

Comparing S100G expression across rat strains and developmental stages requires standardized approaches to account for biological variability:

Strain comparison strategies:

  • Strain selection considerations:

    • Include commonly used laboratory strains (Sprague-Dawley, Wistar, Long-Evans, Fischer)

    • Consider including disease-relevant strains (e.g., spontaneously hypertensive rats)

    • Maintain genetic homogeneity through appropriate breeding strategies

  • Experimental design for strain comparisons:

    • Age and sex matching across strains

    • Standardized housing and environmental conditions

    • Simultaneous sample collection and processing

    • Inclusion of strain-specific reference ranges

  • Data normalization approaches:

    • Use strain-specific reference genes validated for stability across compared strains

    • Consider using absolute quantification with standard curves for direct comparability

    • Include internal control samples processed across all experimental batches

Developmental profiling approaches:

  • Age point selection:

    • Critical developmental transitions (prenatal, neonatal, juvenile, adult, aged)

    • More frequent sampling around periods of known developmental changes

    • Consistent age definitions across studies (specific days post-conception or post-birth)

  • Tissue-specific considerations:

    • Account for changing tissue composition during development

    • Consider normalized expression to cell-specific markers

    • Include histological analysis to correlate with tissue development

  • Longitudinal vs. cross-sectional approaches:

    • Cross-sectional: Different animals at each age point (higher animal numbers, no repeated measures bias)

    • Longitudinal: Same animals followed over time (reduced animal numbers, potential for repeated sampling effects)

Analytical considerations:

  • Statistical approaches:

    • Mixed-effects models for longitudinal data

    • ANOVA with post-hoc tests for cross-sectional comparisons

    • Regression analysis for identifying developmental trends

  • Visualization methods:

    • Growth curve modeling to characterize developmental trajectories

    • Heat maps for visualizing expression across multiple tissues and time points

    • Principal component analysis to identify patterns in multidimensional data

Integration with functional assessments:

  • Correlate expression changes with functional measures (e.g., calcium handling capacity)

  • Consider parallel analysis of related genes to identify compensatory mechanisms

  • Include functional validation of significant developmental transitions

What is the role of S100G in rat models of calcium-related disorders beyond pancreatitis?

S100G plays important roles in multiple calcium-related disorders beyond pancreatitis in rat models:

Intestinal disorders:

  • Inflammatory bowel disease models:

    • S100G expression is altered in experimental colitis

    • May function as a protective factor by buffering intracellular calcium fluctuations

    • Expression changes correlate with disease severity and progression

    • S100G induction may represent a compensatory mechanism during intestinal inflammation

  • Malabsorption syndromes:

    • S100G is crucial for intestinal calcium absorption

    • Expression decreases in vitamin D deficiency models

    • Alterations in S100G contribute to calcium malabsorption phenotypes

    • Therapeutic targeting of S100G pathways may improve calcium absorption

Neurological conditions:

  • Neurodegenerative models:

    • Altered S100G expression in specific neuronal populations

    • May contribute to calcium dysregulation in vulnerable neurons

    • Expression patterns differ between "persistently S100-positive" and "variably S100-positive" neurons during neurodegeneration

  • Cerebral ischemia:

    • S100G may serve protective functions during ischemic events

    • Expression changes correlate with neuronal vulnerability

    • Potential target for neuroprotective interventions

Kidney disorders:

  • Renal calcium handling disorders:

    • S100G participates in renal calcium reabsorption

    • Expression alterations contribute to hypercalciuria phenotypes

    • Compensatory upregulation occurs in response to calcium challenges

Bone metabolism disorders:

  • Osteoporosis models:

    • S100G expression correlates with calcium mobilization from bone

    • Expression changes during altered bone remodeling

    • Potential contributor to calcium homeostasis during bone loss

Research approach considerations:

  • Studies should examine tissue-specific expression patterns across multiple disease models

  • Functional consequences should be assessed through calcium imaging and physiological measurements

  • Genetic manipulation approaches can help establish causality rather than correlation

  • Therapeutic interventions targeting S100G should be evaluated for tissue-specific effects

How does S100G expression respond to physiological challenges such as dietary calcium restriction in rat models?

S100G expression demonstrates dynamic responses to physiological challenges, particularly dietary calcium restriction:

Response patterns to dietary calcium restriction:

  • Intestinal adaptation:

    • Upregulation of S100G in intestinal epithelial cells

    • Enhanced calcium absorption efficiency

    • Coordination with vitamin D-dependent mechanisms

    • Increased expression correlates with calcium transport capacity

  • Renal adaptation:

    • Modified S100G expression in renal tubules

    • Enhanced calcium reabsorption to minimize urinary calcium loss

    • Coordinated regulation with other calcium transporters

  • Temporal response characteristics:

    • Rapid initial response (within days)

    • Sustained adaptation with prolonged restriction

    • Return to baseline upon calcium repletion

Regulatory mechanisms:

  • Vitamin D-dependent regulation:

    • 1,25-dihydroxyvitamin D3 is a primary regulator of S100G expression

    • Activation of vitamin D receptor elements in the S100G promoter

    • Increased circulating 1,25-dihydroxyvitamin D3 during calcium restriction

  • Vitamin D-independent mechanisms:

    • Direct calcium-sensing mechanisms

    • Hormonal regulation via parathyroid hormone

    • Local tissue factors and inflammatory mediators

Compensatory mechanisms:

In knockout models, the absence of S100G triggers compensatory upregulation of other calcium transport mechanisms. Studies have shown that Calbindin-D9k knockout mice show no significant phenotype in serum calcium levels due to these compensatory mechanisms, which include increased expression of other calcium transporters and enhanced vitamin D signaling .

Experimental design considerations:

When studying S100G responses to calcium restriction, researchers should:

  • Include time-course measurements to capture both acute and chronic adaptations

  • Assess tissue-specific responses across multiple calcium-handling tissues

  • Measure both gene and protein expression, as post-transcriptional regulation may occur

  • Include functional measurements of calcium absorption and handling

  • Consider potential sex differences in adaptive responses

What insights have been gained about S100G function from comparative studies across different rodent species?

Comparative studies across rodent species have provided valuable insights into S100G function and evolution:

Cross-species conservation and divergence:

  • Structural conservation:

    • High conservation of protein structure across rodent species

    • S100G amino acid sequence shows approximately 90-95% homology between rats and mice

    • Calcium-binding domains are particularly well-conserved

    • Only minor, conservative amino acid substitutions are typically observed between species

  • Expression pattern differences:

    • Species-specific differences in tissue distribution patterns

    • Varied developmental expression timelines

    • Different responses to physiological challenges across species

Functional implications from comparative studies:

  • Compensatory mechanisms:

    • Different rodent species show varied compensatory responses to S100G deficiency

    • Species-specific redundancy in calcium regulatory pathways

    • Varied reliance on S100G versus other calcium binding proteins

  • Physiological adaptation:

    • Species adaptations reflect evolutionary pressures on calcium homeostasis

    • Dietary calcium requirements and absorption efficiency vary across species

    • Reproductive demands drive species-specific adaptations in calcium regulation

Methodological insights:

  • Model selection considerations:

    • Rat models may be preferred for certain applications based on tissue-specific expression patterns

    • Mouse models offer greater genetic manipulation possibilities

    • Selection should be based on specific research questions and translational goals

  • Translational relevance:

    • Identification of conserved mechanisms across species strengthens translational potential

    • Species-specific differences highlight limitations in extrapolating to human physiology

    • Multi-species approaches provide stronger evidence for fundamental mechanisms

Key comparative findings:

  • S100G protein is highly conserved at the amino acid level, with only four conservative changes observed between rat and bovine S100 alpha sequences

  • Expression patterns show that S100 proteins are not brain-specific but are expressed in numerous tissues across species

  • While protein structure is conserved, regulatory mechanisms and expression patterns show more species-specific variation

  • S100G mRNA exhibits similar size (0.5 Kb) across rat tissues, suggesting consistent mRNA processing across tissues

How can S100G research in rats inform our understanding of calcium signaling networks in human diseases?

S100G research in rats provides valuable translational insights for understanding human calcium signaling networks and disease mechanisms:

Translational relevance of rat S100G studies:

  • Structural and functional conservation:

    • High conservation of S100G protein structure between rats and humans

    • Similar tissue distribution patterns across species

    • Conserved calcium-binding mechanisms and affinities

    • Comparable regulatory pathways controlling expression

  • Disease modeling applications:

    • Rat models of calcium-related disorders recapitulate key features of human pathology

    • S100G dysregulation patterns in rat disease models often parallel human conditions

    • Therapeutic interventions targeting S100G pathways may have human applications

Specific translational insights:

  • Pancreatitis mechanisms:

    • S100G induction in acinar cells during pancreatitis represents a conserved response

    • Calcium buffering function may be protective in both rat and human pancreatitis

    • Potential biomarker application for disease progression and severity

  • Intestinal calcium absorption:

    • Fundamental mechanisms of calcium transport involving S100G are conserved

    • Regulatory pathways responding to vitamin D are similar across species

    • Compensatory mechanisms in deficiency states follow comparable patterns

  • Neurological conditions:

    • Neuron-specific expression patterns provide insights into human brain calcium regulation

    • Protective roles in neuronal calcium homeostasis may be relevant to human neuropathology

    • Understanding "persistently S100-positive" versus "variably S100-positive" neuronal populations has implications for human brain research

Research strategy recommendations:

  • Complementary approach:

    • Use rat models for in-depth physiological and molecular studies

    • Validate key findings in human samples when available

    • Employ comparative genomics to strengthen translational relevance

  • Focus areas with highest translational potential:

    • Therapeutic targeting of S100G pathways in calcium-related disorders

    • Biomarker development for disease progression and treatment response

    • Mechanistic understanding of calcium dysregulation in disease states

  • Integration with human data:

    • Correlate rat findings with human genetic association studies

    • Compare expression patterns between rat models and human tissue samples

    • Validate in human cell culture systems when possible

What approaches are recommended for integrating S100G studies with broader calcium signaling research in rat models?

Integrating S100G studies with broader calcium signaling research requires strategic methodological approaches:

Integration strategies:

  • Multi-protein analysis approaches:

    • Simultaneous profiling of S100G alongside other calcium binding proteins

    • Analysis of complete calcium signaling networks rather than isolated components

    • Investigation of compensatory mechanisms within calcium regulatory systems

  • Multi-level integration:

    • Connect genomic, transcriptomic, proteomic, and functional data

    • Link molecular findings to cellular, tissue, and whole-organism phenotypes

    • Establish cause-effect relationships across biological scales

  • Pathway mapping approaches:

    • Place S100G within comprehensive calcium signaling maps

    • Identify convergent and divergent pathways

    • Characterize pathway redundancy and robustness

Methodological recommendations:

  • Combined technical approaches:

    • Calcium imaging paired with molecular detection methods

    • Real-time monitoring of multiple calcium regulatory proteins

    • Simultaneous manipulation of multiple pathway components

  • Systems biology approaches:

    • Mathematical modeling of calcium signaling networks including S100G

    • Prediction of system behavior under varying conditions

    • Identification of critical nodes and feedback mechanisms

  • Multi-tissue analysis:

    • Comparative analysis across calcium-regulated tissues

    • Identification of tissue-specific versus universal mechanisms

    • Understanding of coordinated calcium homeostasis across organ systems

Key research questions to address:

  • How does S100G interact with calcium channels, pumps, and exchangers?

  • What is the relationship between S100G and other calcium-binding proteins?

  • How do S100G-mediated processes integrate with other calcium-dependent signaling pathways?

  • What compensatory mechanisms engage when S100G function is altered?

Experimental design frameworks:

  • Include comprehensive analysis of multiple calcium regulatory proteins in any S100G study

  • Design experiments to specifically test for functional redundancy

  • Consider calcium signaling as a dynamic network rather than linear pathways

  • Employ both genetic and pharmacological approaches to manipulate pathway components

How can researchers effectively apply multi-omics approaches to understand S100G regulation in rat models?

Multi-omics approaches offer powerful strategies for comprehensively understanding S100G regulation:

Integrated multi-omics workflow:

  • Genomic analysis:

    • Identification of genetic variants affecting S100G expression

    • Analysis of promoter and enhancer regions

    • Characterization of transcription factor binding sites

    • Investigation of epigenetic modifications (DNA methylation, histone modifications)

  • Transcriptomic approaches:

    • RNA-Seq for comprehensive gene expression profiling

    • Small RNA sequencing to identify miRNA regulators of S100G

    • Alternative splicing analysis

    • Long non-coding RNA characterization

  • Proteomic strategies:

    • Global proteome profiling to identify co-regulated proteins

    • Post-translational modification analysis of S100G

    • Protein-protein interaction mapping

    • Protein turnover and stability assessment

  • Metabolomic integration:

    • Analysis of calcium-related metabolites

    • Correlation of metabolic state with S100G expression

    • Investigation of vitamin D metabolites and related compounds

Data integration frameworks:

  • Computational approaches:

    • Network analysis to identify regulatory hubs

    • Pathway enrichment analysis across multiple omics datasets

    • Machine learning for pattern recognition and predictive modeling

    • Bayesian network analysis for causal relationship inference

  • Visualization strategies:

    • Multi-layered network visualization

    • Integrated heatmaps across data types

    • Temporal and spatial mapping of multi-omics data

  • Validation approaches:

    • Targeted experimental validation of key predictions

    • Iterative modeling with experimental feedback

    • Cross-validation across multiple experimental models

Application examples:

An integrated multi-omics approach applied to S100G research might reveal:

  • Transcription factors regulating S100G expression in different tissues

  • miRNAs providing post-transcriptional regulation

  • Protein interaction partners modulating S100G function

  • Metabolic conditions influencing S100G activity

Practical implementation considerations:

  • Begin with hypothesis-generating global profiling

  • Follow with targeted validation of key findings

  • Consider both steady-state and dynamic response analyses

  • Include appropriate bioinformatics expertise in research teams

  • Develop clear data management plans for large-scale data integration

Product Science Overview

Introduction

S100 Calcium Binding Protein G, also known as calbindin-D9k, is a member of the S100 protein family. This family consists of low-molecular-weight proteins characterized by their ability to bind calcium ions. The S100 proteins play crucial roles in various cellular processes, including cell cycle progression, differentiation, and regulation of intracellular calcium levels.

Structure and Function

S100G, or calbindin-D9k, is a small cytosolic protein with a molecular weight of approximately 9 kDa. It contains two EF-hand motifs, which are helix-loop-helix structures capable of binding calcium ions. The binding of calcium induces conformational changes in the protein, which are essential for its function.

Expression and Localization

S100G is predominantly expressed in the intestine, where its expression is regulated by vitamin D. The protein is involved in the active transport of calcium across the intestinal epithelium, playing a critical role in calcium homeostasis. Additionally, S100G is found in other tissues, including the kidneys and placenta, where it may have similar functions related to calcium transport and regulation.

Recombinant Production

Recombinant S100G (Rat) is produced using molecular cloning techniques. The gene encoding the rat S100G protein is inserted into an expression vector, which is then introduced into a host organism, typically Escherichia coli. The host cells express the protein, which is subsequently purified using various chromatographic techniques. Recombinant production allows for the generation of large quantities of the protein for research and therapeutic purposes.

Research and Applications

S100G has been extensively studied for its role in calcium metabolism and its potential implications in various diseases. Research has shown that alterations in S100G expression are associated with conditions such as osteoporosis, chronic kidney disease, and certain cancers. The protein is also used as a marker in studies investigating calcium absorption and metabolism.

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.