Resistin Mouse, Flag

Resistin Mouse Recombinant, Flag Tag
Shipped with Ice Packs
In Stock

Description

Functional Roles in Metabolic Regulation

Resistin Mouse, Flag has been implicated in:

  • Insulin resistance: Reduces insulin-stimulated glucose uptake in skeletal muscle, liver, and adipose tissue .

  • Lipid metabolism: Promotes lipolysis in adipose tissue, increasing serum free fatty acids (FFA) and glycerol .

  • Adipogenesis: Inhibits differentiation of adipocytes in murine models .

Mechanisms:

  • Attenuates insulin signaling via reduced tyrosine phosphorylation of IRS-1 and Akt in muscle and adipose tissue .

  • Induces pro-inflammatory cytokines (e.g., TNF-α, MCP-1) in macrophages, exacerbating adipose tissue inflammation .

Table 1: Experimental Insights from Resistin Mouse, Flag Studies

Study FocusMethodologyKey OutcomeSource
Insulin SensitivityHyperinsulinemic-euglycemic clamp39% lower glucose infusion rate (GIR) in transgenic mice vs. controls
Glucose UptakeInsulin tolerance tests56% reduction in skeletal muscle glucose uptake
LipolysisEx vivo adipose tissue analysisIncreased basal lipolysis and reduced insulin suppression
Inflammatory ResponseLPS-treated macrophagesUpregulation of TNF-α, ICAM-1, and VCAM-1 in humanized resistin mice

Controversies and Limitations

  • Obesity paradox: While some studies associate elevated resistin with obesity-induced insulin resistance , others report suppressed resistin expression in obese rodents .

  • Species specificity: Mouse resistin is adipocyte-derived, whereas human resistin is primarily macrophage-derived, complicating translational relevance .

  • Dimerization: FLAG-tagged resistin forms disulfide-linked dimers (Cys26), though biological activity may not require multimerization .

Applications in Research

  • Mechanistic studies: Used to dissect insulin resistance pathways in murine models .

  • Therapeutic screening: Evaluates compounds targeting metabolic inflammation .

  • Protein interaction assays: FLAG tag enables immunoprecipitation and Western blotting .

Product Specs

Introduction

Resistin, encoded by the RSTN gene, is a peptide hormone classified as a cysteine-rich secreted protein within the RELM family. It is also known as ADSF (Adipose Tissue-Specific Secretory Factor) and FIZZ3 (Found in Inflammatory Zone). Human resistin, initially a 108-amino acid prepeptide, undergoes cleavage of its hydrophobic signal peptide before secretion. In human blood, it circulates as a dimer, formed by two 92-amino acid polypeptides linked by a disulfide bond at Cys26.
Resistin is suggested to play a role in obesity resistance. Studies indicate that mouse resistin, primarily produced and secreted by adipocytes, can decrease the sensitivity of skeletal muscle myocytes, hepatocytes, and adipocytes to insulin. Elevated resistin levels have been observed in obese mice, with levels decreasing during fasting and upon administration of antidiabetic drugs. Conversely, Way et al. reported that resistin expression is significantly reduced in obesity and upregulated by certain antidiabetic drugs.
Additional research suggests that mouse resistin levels increase during adipocyte differentiation while simultaneously inhibiting the process. In contrast, human adipogenic differentiation appears to correlate with downregulation of resistin gene expression.

Description
The Resistin Mouse protein is engineered with a phage fd signal sequence (21 amino acids) and a C-terminal flagTag (10 amino acids). This results in a 13.7 kDa recombinant fusion protein comprising 93 amino acids of the Resistin Mouse sequence and 31 additional amino acids from the phage fd signal sequence and the flagTag (underlined).
Physical Appearance
White lyophilized (freeze-dried) powder that has been filtered.
Formulation
The product is filtered (0.4 µm) and lyophilized from a 0.5 mg/ml solution in 0.05M Acetate buffer with a pH of 4.
Solubility
To create a working stock solution of approximately 0.5 mg/mL, we recommend adding 0.1M Acetate buffer (pH 4) to the lyophilized pellet and allowing it to fully dissolve. For use at a higher pH, dilute the solution to a concentration of 10 µg/ml using the appropriate buffer. This antigen exhibits limited solubility at higher concentrations. Note that this product is not sterile. Before using in cell culture, it must be filtered with an appropriate sterile filter.
Stability
The lyophilized protein should be stored 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 has shown no changes after two weeks at this temperature.
Purity
The purity is greater than 95% as determined by SDS-PAGE analysis.
Synonyms
Cysteine-rich secreted protein FIZZ3, Adipose tissue-specific secretory factor, ADSF, C/EBP-epsilon-regulated myeloid-specific secreted cysteine-rich protein, Cysteine-rich secreted protein A12-alpha-like 2, RSTN, XCP1, RETN1, MGC126603, MGC126609.
Source
Escherichia Coli.
Amino Acid Sequence
MKKLLFAIPL VVPFYSHSTM ASMPLCPIDE AIDKKIKQDF NSLFPNAIKN IGLNCWTVSS RGKLASCPEG TAVLSCSCGS ACGSWDIREE KVCHCQCARI DWTAARCCKL QVASLEDYKD DDDK.

Q&A

What is mouse resistin and how does it differ from human resistin?

Mouse resistin is a cysteine-rich protein primarily expressed in mature white adipocytes in rodents. The critical difference between mouse and human resistin lies in their cellular expression patterns and sequence homology. Mouse and human resistin share approximately 60% sequence identity, which is relatively low for hormones conserved across species . While mouse resistin is predominantly expressed by adipocytes, human resistin is primarily produced by macrophages and other inflammatory cells, not adipocytes .

This fundamental difference in expression patterns creates significant challenges when translating research findings from murine models to human applications. The divergence appears to be attributable to the loss of a genomic binding site for the nuclear receptor PPARγ in humans, which controls adipocyte-specific expression of the mouse resistin gene . Despite these differences, both mouse and human resistin levels decrease with thiazolidinedione treatment, suggesting some conserved regulatory mechanisms .

What is the significance of FLAG-tagged mouse resistin in research applications?

FLAG-tagged mouse resistin represents a valuable research tool that enables precise tracking, purification, and functional analysis of the protein in experimental settings. The FLAG epitope tag is a small peptide sequence fused to the N-terminus of the mature resistin peptide (amino acids 21-114), allowing for specific detection using anti-FLAG antibodies .

The recombinant FLAG-tagged mouse resistin is typically expressed in mammalian cell lines such as HEK293 cells and purified from conditioned media . This approach offers several advantages:

  • Enhanced detection sensitivity in complex biological samples

  • Simplified purification through affinity chromatography

  • Ability to distinguish exogenous from endogenous resistin

  • Consistent protein quality for reproducible experimental outcomes

When designing experiments with FLAG-tagged resistin, researchers should consider that the tag might potentially influence protein folding, oligomerization, or receptor interactions, necessitating appropriate controls with untagged protein when possible.

How does resistin expression vary under different physiological conditions in mice?

Mouse resistin expression demonstrates significant responsiveness to various physiological and pathological states:

ConditionEffect on Resistin ExpressionEffect on Circulating Levels
FastingDecreased in adipose tissueReduced in serum
Refeeding after fastTends to increaseReturns toward baseline
Diet-induced obesitySuppressed mRNA in adipose tissueElevated in circulation
Genetic obesity (ob/ob mice)Reduced mRNA expressionIncreased in serum
TZD drug treatmentReduced expressionDecreased levels
High glucose conditionsUpregulated expressionIncreased levels
Inflammatory cytokines (e.g., TNF-α)Suppressed expressionVariable response

These seemingly contradictory findings between tissue expression and circulating levels in obesity models suggest complex post-transcriptional regulation and potentially increased resistin half-life or altered clearance mechanisms in obese states . Researchers should carefully consider these variables when designing studies examining resistin biology under various metabolic conditions.

What is the current understanding of mouse resistin's receptor interactions?

Recent research has identified ROR1 (receptor tyrosine kinase-like orphan receptor 1) as a potential functional receptor for mouse resistin . This interaction appears to be specific to defined domains of the ROR1 receptor:

Mouse resistin specifically interacts with the Frizzled-like (FZZ-like) and Kringle (KR) domains of the ROR1 extracellular region . This interaction results in inhibition of ROR1 tyrosine phosphorylation, suggesting that resistin may function as an inhibitory ligand for this receptor .

The resistin-ROR1 interaction has significant downstream effects on cellular signaling pathways, including:

  • Modulation of ERK1/2 phosphorylation

  • Regulation of suppressor of cytokine signaling 3 (SOCS3) expression

  • Altered glucose transporter 4 (GLUT4) and glucose transporter 1 (GLUT1) expression

This discovery represents a significant advancement in understanding resistin's molecular mechanisms, as previous research had not clearly identified specific cellular receptors. The cysteine-rich nature of both resistin and ROR1 likely facilitates this protein-protein interaction, as many cysteine-rich proteins function as receptor-ligand pairs .

How should researchers reconcile contradictory findings on resistin's role in adipogenesis?

The literature contains seemingly contradictory findings regarding resistin's effects on adipogenesis. Some studies suggest resistin inhibits adipogenesis, while others indicate it promotes adipocyte differentiation . To reconcile these contradictions, researchers should consider:

  • Experimental timing: The effects of resistin may vary depending on the stage of adipocyte differentiation at which it is applied.

  • Concentration dependence: Different resistin concentrations may exert opposite effects, suggesting dose-response studies are essential.

  • Model systems: The specific cell line or primary cell type used (e.g., 3T3-L1 versus primary preadipocytes) may respond differently to resistin.

  • Receptor expression: Variation in ROR1 or other potential resistin receptor expression across different experimental models may explain divergent outcomes.

  • Experimental methodology: Recent evidence indicates that resistin promotes adipogenesis of 3T3-L1 cells through ROR1-dependent mechanisms . Researchers should employ genetic approaches (siRNA knockdown or CRISPR/Cas9 editing of ROR1) to definitively establish receptor-dependence of observed effects.

A comprehensive experimental approach should include multiple time points, concentration ranges, and both gain- and loss-of-function approaches to fully characterize resistin's role in adipogenesis.

What methodological approaches are recommended for studying resistin's effects on glucose homeostasis?

When investigating resistin's impact on glucose homeostasis, researchers should implement multiple complementary approaches:

In vitro models:

  • Glucose uptake assays in adipocytes, skeletal muscle cells, and hepatocytes using radiolabeled glucose

  • Western blotting to assess insulin signaling pathway components (IRS-1, Akt, AS160 phosphorylation)

  • qRT-PCR analysis of gluconeogenic enzymes in hepatocytes

  • GLUT4 translocation assays using fluorescently-tagged GLUT4 constructs

In vivo models:

  • Glucose tolerance tests (GTT) and insulin tolerance tests (ITT) in wild-type, resistin knockout, and resistin-overexpressing mice

  • Hyperinsulinemic-euglycemic clamp studies to assess insulin sensitivity

  • Tissue-specific glucose uptake using 2-deoxyglucose

  • Measurement of hepatic glucose production using isotope dilution techniques

The humanized resistin mouse model developed by Qatanani et al. provides a valuable tool for understanding human resistin biology in vivo . This model expresses human resistin via a macrophage promoter in mice lacking endogenous resistin, replicating the human expression pattern. Studies have shown these mice develop white adipose tissue inflammation and insulin resistance, supporting human resistin's role in linking inflammatory responses to glucose homeostasis .

What expression systems are optimal for producing recombinant mouse resistin?

The choice of expression system for recombinant mouse resistin production significantly impacts protein quality and experimental outcomes. Based on published research, the following systems have been successfully employed:

Expression SystemAdvantagesLimitationsApplications
HEK293 cellsMammalian post-translational modifications; secretion into media; FLAG-tag compatibilityHigher cost; lower yieldFunctional studies requiring native structure
E. coliHigh yield; cost-effective; simple purificationLacks glycosylation; refolding often requiredStructural studies; antibody production
Baculovirus/insect cellsIntermediate yield; some post-translational modificationsMore complex than bacterial systemsCompromise between yield and structure

The HEK293 mammalian expression system appears to be preferred for producing biologically active FLAG-tagged mouse resistin . This system enables proper folding and secretion of resistin into the culture medium, facilitating subsequent purification. The recombinant protein should encode the mature peptide (amino acids 21-114) with the FLAG tag positioned at the N-terminus to avoid interfering with the C-terminal disulfide bond formation critical for resistin's oligomerization and activity .

How can researchers distinguish between direct and indirect effects of resistin in complex systems?

Differentiating direct from indirect effects of resistin represents a significant challenge in metabolic research. To address this, researchers should employ a multi-faceted approach:

  • Time-course experiments: Direct effects typically occur more rapidly than indirect effects. Short time courses (minutes to hours) versus long time courses (hours to days) can help distinguish these mechanisms.

  • Receptor-blocking approaches: Utilizing ROR1 neutralizing antibodies or small molecule inhibitors can help determine if observed effects are directly mediated through this receptor .

  • Conditioned media experiments: Comparing direct application of resistin to cells versus conditioned media from resistin-treated cells can help identify effects mediated by secondary factors.

  • Transcriptional/translational inhibitors: Using cycloheximide or actinomycin D to block protein synthesis or transcription, respectively, can determine if resistin's effects require new protein production.

  • Single-cell analysis: Techniques like single-cell RNA-seq can reveal heterogeneous responses within populations and identify direct responder cells.

  • Ex vivo tissue explants: Comparing effects in isolated tissues versus in vivo responses can help discriminate direct tissue effects from systemic adaptations.

Given the inflammatory effects of resistin, particularly human resistin, researchers must carefully design experiments to distinguish primary metabolic effects from those secondary to inflammatory pathway activation .

What are the key considerations for translating mouse resistin research to human applications?

Translating findings from mouse resistin research to human applications requires careful consideration of several critical factors:

  • Different expression patterns: Mouse resistin is primarily expressed in adipocytes, while human resistin is predominantly expressed in macrophages and inflammatory cells . This fundamental difference may result in distinct biological contexts and mechanisms of action.

  • Sequence divergence: The approximately 60% sequence homology between mouse and human resistin suggests potentially different binding partners and signaling pathways.

  • Receptor interactions: Whether human resistin interacts with ROR1 as mouse resistin does remains unclear. Recent evidence suggests human resistin may interact with Toll-like receptor 4 (TLR4) , potentially mediating its proinflammatory effects.

  • Humanized mouse models: Utilizing transgenic mice expressing human resistin from macrophages while lacking endogenous mouse resistin provides a valuable translational tool .

  • Inflammatory context: Human resistin's role in inflammatory processes should be considered when designing translational studies, as it is robustly induced by proinflammatory stimuli like LPS, TNF-α, IL-6, and IL-1β .

  • Clinical correlations: Triangulating findings with human clinical data on resistin levels in various metabolic and inflammatory conditions can strengthen translational relevance.

The development of humanized resistin mouse models has provided important insights, showing that human resistin can indeed contribute to adipose tissue inflammation and insulin resistance in vivo . Future research should continue to develop better models to bridge the gap between mouse and human resistin biology.

How can researchers address issues with resistin stability in experimental settings?

Resistin stability represents a significant challenge in experimental settings. To optimize stability and experimental reproducibility, researchers should consider:

  • Storage conditions: Recombinant FLAG-tagged mouse resistin should be stored in working aliquots at -20°C to avoid repeated freeze-thaw cycles that can degrade the protein .

  • Buffer optimization: Phosphate-buffered saline at pH 7.2 with potential stabilizing agents (0.1% BSA or low concentrations of glycerol) can improve stability .

  • Quality control metrics: Regular assessment of protein purity (>95% by SDS-PAGE) and endotoxin levels (<0.1EU/μg) is essential for reproducible experiments .

  • Oligomerization state: Mouse resistin forms oligomers that may be important for its biological activity. Experimental conditions that disrupt oligomerization should be avoided.

  • Carrier proteins: Adding inert carrier proteins to very dilute resistin solutions can prevent non-specific binding to tubes and loss of protein.

  • Fresh preparation: For critical experiments, using freshly thawed aliquots rather than stored working solutions is recommended.

  • Biological validation: Confirming bioactivity using established assays (e.g., effects on glucose uptake in 3T3-L1 adipocytes) before commencing complex experiments.

By implementing these strategies, researchers can maintain resistin stability for up to three months without detectable loss of activity .

What controls are essential in FLAG-tagged resistin experiments?

Proper controls are critical for interpreting experiments using FLAG-tagged mouse resistin. Essential controls include:

  • Tag-only control: A soluble FLAG peptide or irrelevant FLAG-tagged protein to control for non-specific effects of the tag itself.

  • Denatured protein control: Heat-inactivated FLAG-tagged resistin to distinguish effects requiring native protein structure.

  • Dose-response analysis: Multiple concentrations of resistin to establish dose-dependent effects and physiological relevance.

  • Endotoxin control: LPS contamination can confound results, particularly in inflammation studies. LPS inhibitors (e.g., polymyxin B) or endotoxin-free preparations are essential.

  • Receptor competition: When studying receptor interactions, unlabeled resistin can be used to compete with FLAG-tagged resistin.

  • Antibody specificity controls: When detecting FLAG-tagged resistin by immunological methods, isotype controls and blocking peptides should be employed.

  • Cell-type specificity: Testing effects across multiple cell types (adipocytes, macrophages, hepatocytes) can help establish specificity of observed responses.

  • Genetic validation: siRNA knockdown or CRISPR/Cas9 editing of potential receptors like ROR1 can confirm receptor-mediated effects.

These controls collectively ensure that observed effects are specifically attributable to resistin's biological activity rather than experimental artifacts.

What are the most promising avenues for future resistin research?

Based on current knowledge gaps and recent discoveries, several research directions hold particular promise:

  • Receptor biology: Further characterization of resistin-ROR1 interactions in mice and identification of definitive human resistin receptors, including validation of TLR4 interactions .

  • Tissue-specific effects: Elucidating resistin's tissue-specific actions beyond adipose tissue, particularly in liver, skeletal muscle, and cardiovascular tissues.

  • Integration with other adipokines: Understanding how resistin functions within the broader network of adipose-derived factors like adiponectin, leptin, and inflammatory cytokines.

  • Therapeutic targeting: Development of resistin antagonists or receptor modulators that may have therapeutic potential in metabolic and inflammatory diseases.

  • Biomarker development: Refinement of resistin as a clinical biomarker for diabetes risk assessment, leveraging prospective studies showing resistin predicts future diabetes development .

  • Mechanistic studies: Further elucidation of downstream signaling pathways, particularly the connection between ROR1 inhibition and glucose metabolism/adipogenesis regulation .

  • Translational models: Development of improved humanized models that better reflect human resistin biology in physiologically relevant contexts.

  • Genetic approaches: Exploration of resistin polymorphisms and their functional consequences, as genetic data suggests approximately 70% of resistin expression variation may be attributed to genetic effects .

The convergence of these research directions will likely yield a more comprehensive understanding of resistin biology and its potential therapeutic implications in metabolic and inflammatory disorders.

Product Science Overview

Introduction

Resistin, also known as adipose tissue-specific secretory factor (ADSF) or found in inflammatory zone 3 (FIZZ3), is a cysteine-rich peptide hormone derived from adipose tissue. In mice, it is encoded by the Retn gene and plays a significant role in various physiological and pathological processes .

Structure and Function

Resistin is a member of the resistin-like molecule (RELM) family, which includes RELMα, RELMβ, and RELMγ. The structure of resistin comprises a carboxy-terminal disulfide-rich β-sandwich “head” domain and an amino-terminal α-helical “tail” segment . In mice, resistin is primarily expressed in adipocytes and has been implicated in insulin resistance and type 2 diabetes .

Recombinant Resistin Production

Recombinant resistin is produced using Escherichia coli (E. coli) expression systems. The recombinant protein contains 93 amino acid residues of mouse resistin and 31 additional amino acid residues, including a signal sequence of phage fd and a C-terminal FLAG tag . The FLAG tag is an artificial peptide sequence (DYKDDDDK) used for protein purification and detection .

FLAG Tag

The FLAG tag is a widely used epitope tag for protein purification and detection. It is highly specific and can be used in various assays, including immunoprecipitation, Western blotting, and immunofluorescence . The FLAG tag’s small size and hydrophilic nature make it less likely to interfere with the protein’s function .

Biological Significance

Resistin has been studied extensively for its role in metabolic and inflammatory diseases. In mice, resistin is associated with obesity and insulin resistance, while in humans, it is considered a pro-inflammatory molecule expressed in immune cells . Resistin’s involvement in chronic inflammatory diseases, metabolic disorders, and cancers highlights its importance as a potential therapeutic target .

Applications

Recombinant mouse resistin with a FLAG tag is used in various research applications, including:

  • Studying insulin resistance: Investigating the molecular mechanisms underlying insulin resistance and type 2 diabetes.
  • Inflammatory response: Exploring resistin’s role in chronic inflammatory diseases and immune response modulation.
  • Protein-protein interactions: Using the FLAG tag for affinity purification and studying interactions with other proteins.

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.