S100G Human

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

Description

Biological Functions

S100G plays critical roles in calcium homeostasis and cellular signaling:

  • Calcium Transport: Facilitates transcellular calcium absorption in intestinal enterocytes by buffering cytoplasmic Ca²⁺ and enhancing ATP-dependent transport via PMCA1 at the basolateral membrane .

  • Vitamin D Regulation: Expression is stimulated by calcitriol (active vitamin D metabolite), though mechanisms remain partially debated .

  • Neuroprotection: Exhibits neurotrophic effects during brain development and serves as a biomarker for fetal brain damage in cord blood .

  • Reproductive Roles: Dynamically regulated in uterine endometrium during pregnancy, suggesting roles in maternal-fetal calcium exchange .

Expression Patterns and Tissue Distribution

S100G is expressed in a tissue-specific manner :

Tissue/Cell TypeExpression LevelFunctional Context
Intestinal EpitheliumHighCalcium absorption and transport
Cartilage/BoneModerateMineralization and skeletal development
Brain (Ameloblasts)LowNeuroprotection and developmental signaling
Uterine EndometriumVariablePregnancy-associated calcium regulation

Absent in human kidney but detected in other species . Single-cell RNA sequencing highlights expression in epithelial and neural lineages .

Pathological Associations

  • Neurological Disorders: Elevated S100G in cord blood correlates with fetal brain injury .

  • Cancer: Altered S100 protein expression (including S100G) is linked to tumor progression, though direct oncogenic mechanisms require further study .

  • Inflammatory Diseases: S100 proteins like S100G interact with receptors (e.g., RAGE) to modulate immune responses .

Therapeutic Targets

  • Recombinant S100G: Used in research to study calcium signaling pathways and develop diagnostic tools .

  • Biomarker Potential: Quantification in biological fluids may aid in monitoring neurodevelopmental or pregnancy complications .

Research Challenges and Future Directions

  • Mechanistic Gaps: The interplay between vitamin D and S100G in non-intestinal tissues remains unclear .

  • Therapeutic Development: Neutralizing antibodies or small-molecule inhibitors targeting S100 proteins are under exploration for inflammatory and neoplastic diseases .

Product Specs

Introduction
S100G, also known as calbindin D9K, is a calcium-binding protein regulated by vitamin D. As a member of the calcium-binding protein family, which includes calmodulin, parvalbumin, troponin C, and S100 protein, S100G is found in the cytosol. Its expression in the intestine is influenced by vitamin D and correlates with calcium transport activity. S100G is believed to enhance Ca2+ absorption by buffering intracellular Ca2+ levels and promoting ATP-dependent Ca2+ transport across the duodenal basolateral membrane.
Description
Recombinant Human S100G, expressed in E. coli, has a molecular weight of 10.04 kDa. It comprises 87 amino acid residues of the human S100G protein and incorporates a 9 amino acid Histidine tag at the N-terminus.
Formulation
S100G is filtered through a 0.4 μm filter and lyophilized to a concentration of 0.5 mg/ml in a buffer of 20mM Tris and 50mM NaCl, at pH 7.5.
Solubility
To prepare a working stock solution, add deionized water to the lyophilized pellet to achieve an approximate concentration of 0.5 mg/ml. Allow for complete dissolution. This product is not sterile. Prior to cell culture use, filter the solution through an appropriate sterile filter.
Stability
Store the lyophilized protein at -20°C. After reconstitution, aliquot the product to minimize repeated freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited period and shows no discernible changes for up to two weeks at this temperature.
Applications
Western blotting.
Synonyms
Protein S100-G, Calbindin-D9k, S100 calcium-binding protein G, Vitamin D-dependent calcium-binding protein intestinal, CABP, S100G, CABP9K, CALB3, S100D, CABP1, MGC138379.
Source
Escherichia Coli.
Amino Acid Sequence
MKHHHHHHAS TKKSPEELKRS TKKSPEELKR IFEKYAAKEG DPDQLSKDEL KLLIQAEFPS LLKGPNTLDD LFQELDKNGD GEVSFEEFQV LVKKISQ.

Q&A

What is the molecular structure of human S100G and how does it differ from other S100 family proteins?

S100G (also known as Calbindin 3, CaBP9K, CABP1) is a monomeric 9 kDa protein containing two calcium-binding motifs: a classical C-terminal EF-hand with a canonical Ca²⁺ binding loop and an S100-specific N-terminal EF-hand with a modified Ca²⁺ binding loop called a "pseudo EF-hand." Unlike many other S100 proteins that form dimers, S100G functions as a monomer. The gene encoding human S100G is located on the X-chromosome (Xq22) and consists of three exons containing four Alu repeats .

The protein's structure enables it to bind calcium with high affinity while maintaining structural stability. This distinguishes it from other S100 family members, which typically function as homo- or heterodimers and have different calcium-binding properties and tissue distributions.

What experimental methods are most reliable for detecting S100G expression in human tissue samples?

For reliable S100G detection in human tissues, researchers should consider a complementary methodological approach:

  • mRNA detection: RT-qPCR offers high sensitivity for S100G mRNA quantification, particularly in small intestine samples where S100G is highly expressed. Primer design should account for the three-exon structure of the gene.

  • Protein detection: Western blotting using monoclonal antibodies against S100G, with positive controls from intestinal tissue. Immunohistochemistry can localize the protein within specific cell types, particularly in enterocytes.

  • Functional assays: Calcium-binding assays using purified S100G can confirm protein activity.

  • Bioinformatic validation: Tools like GEPIA 2 can be used to compare your experimental expression data with established datasets from TCGA and GEx projects .

When designing these experiments, include appropriate controls and account for potential cross-reactivity with other S100 family members due to structural similarities.

How does vitamin D regulate S100G expression in human tissues?

S100G expression is primarily regulated by calcitriol (1,25-dihydroxyvitamin D3), the active metabolite of vitamin D. While the exact mechanism remains somewhat controversial, the following experimental approaches have elucidated the relationship:

  • Promoter analysis: Studies have identified vitamin D response elements (VDREs) in the S100G gene promoter region.

  • Vitamin D receptor (VDR) studies: Knockout models show that mice lacking VDR have reduced, but not absent, S100G expression, suggesting both VDR-dependent and VDR-independent regulation mechanisms .

  • Cell culture models: Intestinal epithelial cell lines treated with calcitriol demonstrate upregulation of S100G transcription within hours of treatment.

  • Chromatin immunoprecipitation (ChIP): This technique has confirmed VDR binding to the S100G promoter region following calcitriol stimulation.

Researchers investigating this relationship should design experiments that account for both direct transcriptional regulation and potential indirect mechanisms involving other calcium-regulating pathways.

What tissue distribution pattern does S100G exhibit in humans compared to other species?

The tissue distribution of S100G shows important species-specific differences that researchers must consider when designing translational studies:

TissueHuman ExpressionOther Species (e.g., rodents)
Small intestineHigh (primarily proximal)High
KidneyNot detected (mRNA)Present
UterusNot detected (mRNA)Present
PlacentaNot detected (mRNA)Present
Teeth (ameloblasts)PresentPresent
BonePresentPresent
CartilagePresentPresent

These species differences highlight why caution is necessary when extrapolating findings from animal models to human conditions. Experimental designs should include human tissue validation whenever possible to confirm relevance of findings from animal models .

How should researchers approach S100G protein purification for in vitro studies?

Purification of S100G for functional studies requires careful consideration of its calcium-binding properties:

  • Expression system selection: Bacterial expression systems (E. coli) with pET vectors containing human S100G cDNA have yielded functional protein. Mammalian expression systems may be preferable when post-translational modifications are critical.

  • Purification strategy:

    • Begin with affinity chromatography (His-tag or calcium-dependent phenyl-Sepharose)

    • Follow with size exclusion chromatography to ensure monomeric state

    • Verify purity using SDS-PAGE and Western blotting

  • Activity verification: Calcium-binding assays (isothermal titration calorimetry or fluorescence-based methods) should confirm functionality.

  • Storage considerations: Store purified protein with defined calcium concentrations to maintain stability.

For structural studies, researchers should consider the impact of calcium saturation on protein conformation and adjust conditions accordingly.

What experimental design considerations are crucial for investigating S100G's role in calcium transport in enterocytes?

Investigating S100G's role in intestinal calcium transport requires sophisticated experimental approaches:

  • Experimental models selection:

    • Cell models: Caco-2 cells (polarized human intestinal cells) allow for apical-to-basolateral transport studies

    • Organoid cultures: Intestinal organoids maintain physiological cell organization

    • In vivo models: Consider intestine-specific S100G knockout/knockdown models

  • Control variables: Experimental designs must control for:

    • Vitamin D status (calcitriol levels)

    • Expression of other calcium transport proteins (TRPV6, PMCA1)

    • Calcium concentration gradients

    • Cell polarization status

  • Measurement approaches:

    • Radioactive calcium (⁴⁵Ca) flux assays

    • Fluorescent calcium indicators for real-time imaging

    • Patch-clamp electrophysiology for channel activity

  • Between-subjects vs. within-subjects design:

    • For cell studies, a randomized block design accounting for passage number is recommended

    • For animal studies, littermates should be used to minimize genetic variation

Researchers should apply a combination of gain-of-function (overexpression) and loss-of-function (siRNA knockdown, CRISPR knockout) approaches to establish causality rather than mere correlation .

How can researchers effectively analyze S100G expression data in cancer contexts using bioinformatics tools?

S100G analysis in cancer contexts requires systematic bioinformatic approaches:

This methodological framework enables researchers to generate hypotheses about S100G's potential role in cancer before proceeding to wet-lab validation studies.

What methodological approaches can resolve conflicting data about S100G's neurotrophic and neuroprotective functions?

Resolving contradictions regarding S100G's neurotrophic/neuroprotective effects requires multifaceted experimental designs:

  • Standardization of experimental conditions:

    • Define precise developmental timepoints when studying neurodevelopment

    • Establish consistent stress/injury models for neuroprotection studies

    • Use identical S100G concentrations across studies

  • Cell-specific analyses:

    • Employ single-cell RNA sequencing to identify specific neuronal populations expressing S100G

    • Use cell type-specific conditional knockout models rather than global knockouts

    • Perform co-localization studies with established neuronal/glial markers

  • Functional readouts:

    • Measure multiple endpoints: calcium signaling, neurite outgrowth, cell survival, electrophysiological parameters

    • Assess both short-term and long-term outcomes following S100G manipulation

    • Implement in vivo behavioral testing when using animal models

  • Mechanistic dissection:

    • Distinguish between calcium-dependent and calcium-independent effects

    • Investigate interactions with other calcium-binding proteins

    • Examine downstream signaling pathways using phosphoproteomics

  • Meta-analysis approaches:

    • Perform systematic reviews of published data

    • Use statistical approaches to account for inter-study variability

    • Identify moderating variables that explain contradictory results

Researchers should also consider species differences and developmental timing as potential sources of contradictory findings.

What protein-protein interaction networks involve S100G, and how should researchers investigate these relationships?

Investigating S100G's protein interaction network requires systematic approaches:

  • Computational prediction methods:

    • Use tools like STRING database to identify potential interactions based on co-expression, text mining, and experimental data

    • Apply GeneMANIA to construct gene-gene interaction networks and identify functionally related genes

    • Search for proteins containing S100-binding motifs using pattern recognition algorithms

  • Experimental validation techniques:

    • Co-immunoprecipitation (Co-IP): Pull down S100G and identify binding partners by mass spectrometry

    • Proximity labeling (BioID, APEX): Identify proteins in close proximity to S100G in living cells

    • Yeast two-hybrid screening: Systematic screening for direct protein-protein interactions

    • Surface plasmon resonance (SPR): Measure binding kinetics between S100G and candidate partners

  • Functional validation:

    • Perform co-localization studies using confocal microscopy

    • Use FRET/BRET to confirm direct interactions in living cells

    • Employ mutagenesis of binding interfaces to confirm specificity

  • Integration with pathway analyses:

    • GO enrichment analysis shows S100G relationships involve calcium ion binding, calcium-dependent protein binding, RAGE receptor binding

    • PPI Enrichment Analysis reveals connections to cornified envelope, cornification, and keratinization pathways

Research should focus particularly on interactions with calcium transport machinery (TRPV6, PMCA1) and potential binding partners in the neurotrophic pathway.

How can researchers accurately assess the impact of S100G genetic variants on protein function and disease susceptibility?

Investigating S100G genetic variants requires a comprehensive experimental pipeline:

  • Variant identification and prioritization:

    • Perform targeted sequencing of S100G in relevant patient cohorts

    • Use bioinformatic tools (PolyPhen-2, SIFT, CADD) to predict functional impacts

    • Prioritize variants in calcium-binding domains or regulatory regions

  • Structural and functional assessment:

    • Generate recombinant proteins containing identified variants

    • Measure calcium-binding properties using isothermal titration calorimetry

    • Assess protein stability through thermal shift assays

    • Determine subcellular localization through fluorescent tagging

  • Cellular phenotyping:

    • Create isogenic cell lines using CRISPR/Cas9 knock-in

    • Measure calcium transport efficiency in enterocyte models

    • Assess impact on vitamin D responsiveness

    • Evaluate effects on cell migration, proliferation, and survival

  • Population-level analyses:

    • Conduct case-control association studies in relevant disease cohorts

    • Perform genotype-phenotype correlation analyses

    • Design longitudinal studies to assess variant impact on disease progression

  • Experimental design considerations:

    • Use appropriate statistical power calculations for genetic association studies

    • Implement randomized block designs that account for demographic and environmental factors

    • Consider both between-subjects (different individuals) and within-subjects (longitudinal) approaches

These methodological approaches enable researchers to establish causality between S100G variants and functional consequences relevant to human health and disease.

How should researchers address the challenge of S100G antibody specificity given the structural similarity among S100 family proteins?

The structural similarity among S100 proteins presents significant challenges for specific antibody-based detection of S100G:

  • Antibody validation strategy:

    • Test antibodies on recombinant S100G alongside other S100 family members

    • Validate using S100G-overexpressing and S100G-knockout models

    • Perform peptide competition assays to confirm epitope specificity

    • Use multiple antibodies targeting different epitopes

  • Complementary approaches:

    • Combine antibody-based detection with mRNA quantification

    • Employ mass spectrometry for unambiguous protein identification

    • Use calcium-binding properties for functional discrimination

  • Controls and standards:

    • Include tissue-specific positive and negative controls known to express or lack S100G

    • Use calibrated recombinant protein standards for quantitative analyses

    • Implement spike-in controls to assess recovery efficiency

  • Reporting standards:

    • Document complete antibody information (catalog number, lot, dilution)

    • Report all validation steps performed

    • Acknowledge potential cross-reactivity limitations

These methodological considerations ensure experimental rigor and reproducibility when studying S100G in complex biological samples .

What experimental design approaches best address the tissue-specific functions of S100G across different human organ systems?

Investigating tissue-specific functions of S100G requires targeted experimental designs:

  • Model system selection:

    • Ex vivo tissue explants: Maintain physiological context while allowing experimental manipulation

    • Tissue-specific primary cells: Isolate cells from relevant tissues (enterocytes, osteoblasts)

    • 3D organoid cultures: Recapitulate tissue architecture and cell-cell interactions

    • Conditional knockout models: Target S100G deletion to specific tissues or cell types

  • Tissue-specific functional readouts:

    • Intestine: Calcium absorption using Ussing chambers or radioactive calcium flux

    • Bone/teeth: Mineralization assays, calcium deposition measurements

    • Nervous system: Neurite outgrowth, calcium imaging, electrophysiology

  • Experimental design considerations:

    • Implement randomized block designs grouping subjects by relevant characteristics

    • Use within-subjects designs when possible to control for individual variability

    • Include appropriate tissue-specific controls

  • Integration approaches:

    • Compare S100G function across tissues using standardized assays

    • Identify tissue-specific binding partners through comparative proteomics

    • Analyze tissue-specific promoter usage and alternative splicing

These approaches enable researchers to differentiate the universal calcium-binding functions of S100G from its tissue-specific roles and regulatory mechanisms.

How can researchers effectively study S100G in the context of the entire S100 protein family network?

Studying S100G within the broader S100 family context requires integrated approaches:

  • Comprehensive expression profiling:

    • Analyze co-expression patterns of all 20 S100 family members across tissues

    • Use single-cell RNA sequencing to identify cell populations co-expressing multiple S100 proteins

    • Perform correlation analyses to identify potential functional relationships

  • Network biology approaches:

    • Construct protein-protein interaction networks using STRING and GeneMANIA

    • Identify hub proteins that interact with multiple S100 family members

    • Map S100G-specific interactions versus family-wide interactions

  • Functional redundancy testing:

    • Perform combinatorial knockdown/knockout experiments

    • Rescue experiments with different S100 family members

    • Identify unique versus overlapping functions

  • Data visualization and integration:

    • Use heatmaps to visualize expression patterns across tissues

    • Create network diagrams showing functional relationships

    • Employ machine learning to identify patterns in large-scale datasets

This systematic approach enables researchers to position S100G within the broader functional landscape of calcium-binding proteins and identify its unique contributions to cellular physiology.

What emerging technologies might advance our understanding of S100G function in calcium homeostasis?

Several cutting-edge technologies hold promise for deepening our understanding of S100G:

  • Advanced imaging techniques:

    • Intravital microscopy: Visualize calcium dynamics and S100G activity in living tissues

    • Super-resolution microscopy: Resolve subcellular localization beyond diffraction limits

    • Calcium biosensors: Monitor real-time calcium flux in specific subcellular compartments

  • Genome editing approaches:

    • CRISPR-Cas9 base editing: Create precise point mutations to study structure-function relationships

    • CRISPR interference/activation: Modulate S100G expression with temporal precision

    • Knock-in reporter systems: Tag endogenous S100G to track expression and localization

  • Single-cell technologies:

    • Single-cell proteomics: Quantify S100G protein levels in individual cells

    • Spatial transcriptomics: Map S100G expression within tissue architecture

    • Multi-omics integration: Correlate S100G expression with epigenetic, transcriptomic, and proteomic data

  • Computational approaches:

    • Molecular dynamics simulations: Model calcium binding and protein-protein interactions

    • Systems biology modeling: Integrate S100G into calcium homeostasis networks

    • Machine learning: Predict novel functions and interactions from large datasets

Researchers should design experiments that leverage these technologies while maintaining rigorous controls and validation strategies to advance the field beyond current limitations .

How should researchers design experiments to investigate the potential role of S100G in disease pathogenesis?

Investigating S100G in disease contexts requires carefully designed experimental approaches:

  • Disease model selection:

    • Patient-derived samples: Analyze S100G expression in affected versus healthy tissues

    • Animal models: Use disease-specific models with S100G manipulation

    • In vitro disease models: Employ relevant stress conditions (inflammation, oxidative stress)

  • Experimental design considerations:

    • Case-control studies: Compare S100G expression/function between disease and control groups

    • Longitudinal designs: Track S100G changes during disease progression

    • Intervention studies: Assess effects of S100G modulation on disease outcomes

  • Functional characterization:

    • Gain/loss-of-function: Overexpress or knock down S100G in disease models

    • Rescue experiments: Determine if restoring S100G function reverses disease phenotypes

    • Pathway analysis: Identify disease-relevant pathways affected by S100G

  • Clinical correlation:

    • Biomarker potential: Evaluate S100G as a diagnostic or prognostic biomarker

    • Genetic association: Analyze S100G variants in disease cohorts

    • Therapeutic targeting: Explore modulation of S100G as a treatment strategy

These approaches enable researchers to establish whether S100G plays a causal role in disease pathogenesis or represents a secondary consequence of disease processes.

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 more than 20 low-molecular-weight calcium-binding proteins characterized by the presence of EF-hand motifs, which are helix-loop-helix structures that bind calcium ions. The S100 proteins play crucial roles in various cellular processes, including cell cycle regulation, differentiation, and signal transduction .

Structure and Function

S100 Calcium Binding Protein G is a cytosolic protein that binds calcium ions in a vitamin D-dependent manner. It is primarily found in the intestine, where its expression correlates with calcium transport activity. The protein has a molecular weight of approximately 10-14 kDa and contains two EF-hand motifs that allow it to bind two calcium ions with different affinities .

The binding of calcium ions induces a conformational change in the protein, exposing hydrophobic regions that facilitate interactions with target proteins. These interactions are essential for the protein’s role in calcium signal transduction and regulation of various cellular processes .

Expression and Regulation

The expression of S100 Calcium Binding Protein G is regulated by several transcription factors, including c-Myc, P53, NF-κB, USF, and Nrf2. The expression level varies depending on the specific cell type and the physical and chemical environment. Additionally, epigenetic modifications such as histone acetylation and DNA methylation also play a role in regulating the expression of this protein .

Role in Disease

Altered expression of S100 Calcium Binding Protein G has been associated with various pathological conditions. For instance, its expression is often dysregulated in cancers, where it may play a role in tumor progression and metastasis. The protein’s involvement in calcium-dependent cellular processes makes it a potential biomarker for differential diagnosis and prognosis evaluation in various diseases .

Human Recombinant Form

The human recombinant form of S100 Calcium Binding Protein G is produced using recombinant DNA technology. This involves inserting the gene encoding the protein into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities. The recombinant protein is then purified and used for various research and therapeutic applications.

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.