Resistin Mutant Human is a modified form of human resistin protein where a critical cysteine residue has been mutated to prevent dimerization. Wild-type human resistin is a 9.9 kDa protein containing 108 amino acids as a prepeptide, whose hydrophobic signal peptide is cleaved before secretion, resulting in a 92-amino acid polypeptide that normally circulates in human blood as a disulfide-linked dimeric protein via Cys26 . In contrast, the mutant form has had this specific cysteine residue altered, preventing the formation of the disulfide bond that enables dimerization, potentially causing it to act as an antagonist to normal resistin function . This structural modification represents a significant research tool as it allows scientists to investigate the specific role of resistin dimerization in its biological functions. The mutation effectively creates a monomeric version of the protein that maintains the primary sequence but exhibits altered tertiary structure and potentially different binding characteristics to receptors and other interaction partners in biological systems .
Human and mouse resistin exhibit profound differences in expression patterns, cellular sources, and sequence homology that impact their comparative biology. While mouse resistin is specifically produced and secreted by adipocytes, human resistin is predominantly expressed in macrophages, with its presence in human adipose tissue attributed mainly to non-adipocyte resident inflammatory cells . The gene and protein sequences share only about 60% identity between humans and rodents, which is less than most hormones conserved across species . Despite these structural differences, the genes are syntenic, with the resistin gene (RETN) located at a similar distance from the Insulin Receptor gene on their respective chromosomes . Functionally, mouse resistin acts on skeletal muscle myocytes, hepatocytes, and adipocytes to reduce insulin sensitivity, whereas human resistin serves primarily as a proinflammatory cytokine . This species-specific divergence creates significant challenges when translating mouse model findings to human disease conditions, necessitating careful consideration of these differences when designing experiments or interpreting results across species .
Recent population-based studies have revealed significant associations between circulating resistin levels, RETN gene variants, and long-term health outcomes. Research from the Taiwan Biobank has demonstrated that participants with progressively increasing resistin levels exhibited higher hazard ratios for all-cause mortality and cancer mortality compared to those with lower resistin levels . Furthermore, individuals with multiple RETN variants (high mutation burden) showed dramatically elevated hazard ratios—3.99 for all-cause mortality and 5.55 for cancer mortality—compared to those without such genetic burden . Genome-wide association studies (GWAS) and DNA methylation analyses have identified specific RETN variants, including rs3219175, rs370006313, and rs3745368, that independently associate with circulating resistin levels, with functional assays confirming that the rs370006313 variant plays a key role in affecting RETN promoter activity . These findings establish both resistin levels and RETN variants as potential biomarkers for predicting long-term outcomes in general populations, particularly in cardiovascular disease and cancer progression risk assessment .
Producing high-quality Resistin Mutant Human protein for research applications requires specific expression systems and purification protocols to ensure biological activity. The most commonly employed expression system is E. coli, which can efficiently produce the 9.9 kDa protein containing 93 amino acid residues . Recombinant Resistin Mutant Human is typically lyophilized from a concentrated (1mg/ml) solution containing 0.1% TFA to maintain stability during storage . The purification process generally involves affinity chromatography, often utilizing histidine tags for selective binding and elution, followed by size exclusion chromatography to separate the monomeric mutant protein from any residual oligomeric forms . When reconstituting the lyophilized protein, researchers should add a small volume of acidified buffer initially before diluting to the desired concentration with the appropriate experimental buffer . Quality control through methods such as HPLC, SDS-PAGE, and mass spectrometry is essential to confirm >99% purity and proper molecular weight, ensuring the protein's experimental reliability for downstream applications in cellular and molecular assays .
Investigating the antagonistic properties of Resistin Mutant Human requires multi-faceted experimental approaches focusing on competitive binding and functional inhibition. Researchers should first establish dose-response curves for wild-type resistin in relevant biological assays, such as insulin signaling pathway activation in target cells, inflammatory cytokine production in macrophages, or monocyte adhesion to endothelial cells . Subsequently, competition assays where increasing concentrations of the mutant protein are added alongside a fixed concentration of wild-type resistin can reveal antagonistic effects through diminished wild-type activity . Cell-based assays using human macrophages, adipocytes, or vascular endothelial cells are particularly valuable, as these are physiologically relevant targets of resistin action . Molecular techniques such as surface plasmon resonance or microscale thermophoresis can quantify binding parameters (KD values) between the mutant resistin and putative resistin receptors, demonstrating whether the mutant competes for the same binding sites as the wild-type protein without triggering downstream signaling . Finally, in vivo models using targeted delivery of Resistin Mutant Human can help evaluate its potential therapeutic applications in conditions characterized by pathological resistin signaling, such as inflammatory states or insulin resistance .
Investigating resistin's chaperone-like activities requires specialized techniques that assess protein folding assistance and protection against denaturation. As demonstrated in previous studies, researchers can employ thermal aggregation assays with heat-labile enzymes such as citrate synthase and Nde1, measuring the ability of human resistin to prevent their aggregation and inactivation under heat stress conditions . Enzyme activity recovery assays provide further insights, where heat or chemically denatured enzymes are incubated with resistin to determine if it can refold and restore their enzymatic functions—a key characteristic of molecular chaperones . Surface hydrophobicity plays a critical role in chaperone activity; therefore, binding assays using hydrophobic probes like Bis-ANS and subsequent function testing can determine the importance of specific hydrophobic regions in resistin's chaperone activity . Site-directed mutagenesis targeting specific residues, such as the Phe49 to Tyr mutation (F49YhRes) previously studied, allows researchers to pinpoint critical amino acids involved in the chaperone function, revealing that while F49YhRes could prevent thermal aggregation, it was unable to refold and restore enzymatic activities . Finally, cellular stress models using tunicamycin or thapsigargin treatment of cell lines like U937 enable the investigation of endogenous resistin localization during endoplasmic reticulum stress and its potential protective roles against apoptosis .
Several significant single nucleotide polymorphisms (SNPs) in the RETN gene have been identified that influence resistin expression levels and disease susceptibility. Among the most extensively studied variants are rs3219175, rs370006313, and rs3745368, which have been independently associated with circulating resistin levels through genome-wide association studies . The functional variant rs370006313 has been demonstrated to directly affect RETN promoter activity through molecular assays . Additional SNPs at positions -638 G>A and -420 C>G in the promoter region have shown strong associations with increased resistin expression in multiple populations . Notably, the minor allele frequencies for -638 G>A have been found to be dramatically lower in Caucasians than Japanese populations, contributing to ethnic variations in resistin expression patterns . The -358 SNP, which is virtually absent in Caucasian populations but present in Japanese individuals, demonstrates the importance of considering ethnic background when studying resistin genetics . These genetic variations collectively account for approximately 70% of the variation in resistin expression levels according to epidemiological studies, highlighting the significant genetic component in determining baseline resistin levels and subsequent disease risk .
Epigenetic regulation of resistin expression involves complex DNA methylation patterns that can be studied through specific methodological approaches. Recent genome-wide DNA methylation analyses have identified CpG sites, particularly cg21271423 and cg09909011, that independently associate with circulating resistin levels . Researchers investigating these epigenetic mechanisms should employ bisulfite conversion techniques followed by pyrosequencing or next-generation sequencing to precisely quantify the methylation status of specific CpG islands in the RETN promoter region . Chromosome conformation capture (3C) assays can reveal how DNA methylation influences long-range chromatin interactions affecting RETN transcription . Analysis of genotype-methylation relationships has demonstrated that certain RETN variants, specifically rs3219175 and rs3745368, show genome-wide significant associations with RETN promoter DNA methylation levels, while others like rs370006313 do not—suggesting different mechanistic pathways of genetic influence on resistin expression . Cell culture models using 5-azacytidine or other demethylating agents can experimentally manipulate methylation states to examine causative relationships between methylation and resistin expression in different cell types . Additionally, chromatin immunoprecipitation (ChIP) assays targeting histone modifications and transcription factor binding at the RETN locus provide further insights into the epigenetic machinery controlling context-specific resistin expression patterns in response to environmental factors such as inflammation or metabolic stress .
Standardizing resistin measurements for clinical research requires addressing several methodological variables to ensure reliable and comparable results across studies. Researchers should establish consensus on sample collection protocols, specifying whether serum or plasma should be used, the appropriate anticoagulants, and standardized fasting conditions prior to blood collection, as postprandial states can influence resistin levels . ELISA-based quantification methods should be validated against reference standards with known concentrations of recombinant human resistin, and laboratories should participate in external quality assessment programs to ensure inter-laboratory consistency . When measuring resistin in clinical samples, researchers must account for potential confounding factors such as concurrent inflammatory conditions, medications (particularly thiazolidinediones which decrease resistin levels), and the time of day when samples are collected due to potential circadian variations . Mass spectrometry-based approaches can provide more precise quantification of specific resistin isoforms, particularly important when studying the relative abundance of monomeric versus oligomeric forms in different disease states . Additionally, establishing population-specific reference ranges is essential given the significant ethnic variations in baseline resistin levels associated with different genetic backgrounds, ensuring that clinical interpretations are made within the appropriate demographic context .
The resistin research field has been marked by significant contradictions that require careful methodological consideration when designing new studies. One fundamental contradiction emerges from species differences—while mouse resistin is predominantly expressed in adipocytes and clearly linked to insulin resistance, human resistin is primarily produced by macrophages and functions as a proinflammatory cytokine . This species-specific divergence necessitates caution when extrapolating findings from mouse models to human disease . Additionally, contradictory findings exist regarding resistin's relationship with obesity; Steppan et al. suggested resistin is elevated in obese mice and down-regulated by fasting and antidiabetic drugs, while Way et al. found that resistin expression is severely suppressed in obesity and stimulated by several antidiabetic drugs . These contradictions highlight the importance of clearly defining experimental conditions, specifying the exact animal models or human populations studied, and carefully controlling for confounding factors such as inflammatory status, medication use, and genetic background . When designing studies to resolve these discrepancies, researchers should consider multi-modal approaches that simultaneously measure resistin levels, gene expression, signaling pathway activation, and functional outcomes in the same experimental system, while also accounting for potential post-translational modifications and oligomeric states of resistin that may affect its biological activity .
Resistin Mutant Human offers several strategic applications in drug discovery pipelines targeting inflammatory and metabolic conditions. As a potential antagonist to wild-type resistin, the mutant protein can serve as a prototype therapeutic molecule for high-throughput screening assays, helping identify small molecules or peptides that mimic its antagonistic effects . Researchers can develop cell-based assays using macrophages, adipocytes, or vascular cells treated with wild-type resistin to induce inflammatory responses or insulin resistance, then screen compound libraries for molecules that mimic the inhibitory effects of Resistin Mutant Human on these pathological processes . Structure-activity relationship studies comparing the mutant and wild-type proteins can identify critical binding interfaces for drug targeting, with crystallography or cryo-EM analysis of the resistin-receptor complex providing atomic-level insights for rational drug design . The mutant protein's ability to prevent resistin oligomerization suggests a therapeutic strategy of disrupting resistin quaternary structure, leading to development of peptide fragments or small molecules that specifically interfere with oligomer formation . Additionally, researchers can explore the therapeutic potential of targeting resistin's novel chaperone-like activity, potentially developing compounds that modulate this function in endoplasmic reticulum stress conditions associated with inflammatory and metabolic disorders .
Investigating resistin's influence on cell signaling pathways requires comprehensive methodological approaches that capture both immediate signaling events and downstream functional consequences. Phospho-specific Western blotting targeting key signaling nodes such as NF-κB, MAPK/ERK, JNK, and Akt pathways provides direct evidence of resistin's activation of inflammatory and insulin-related signaling cascades in target cells like macrophages, vascular cells, and metabolic tissues . Researchers should employ time-course experiments ranging from minutes to hours to capture both rapid phosphorylation events and delayed transcriptional responses, using purified recombinant human resistin at physiologically relevant concentrations (typically 10-100 ng/ml) . RNA sequencing or targeted RT-qPCR arrays can identify transcriptional networks activated by resistin, particularly focusing on inflammatory mediators such as IL-6, TNF-α, and cell adhesion molecules like VCAM-1, ICAM-1, and MCP-1 that are known resistin targets . Pharmacological pathway inhibitors and siRNA knockdown approaches help establish causality in resistin signaling, while CRISPR-Cas9 gene editing can create cellular models lacking specific pathway components to definitively establish their requirement in resistin signal transduction . Finally, the use of Resistin Mutant Human as a competitive inhibitor in these assay systems can reveal which signaling effects depend on oligomerization or specific structural features of wild-type resistin, providing insights into structure-function relationships that inform therapeutic targeting strategies .
The dual functionality of resistin as both a chaperone-like protein and proinflammatory cytokine represents an intriguing molecular intersection that may explain its complex role in disease pathophysiology. During cellular stress conditions such as those induced by tunicamycin or thapsigargin, human resistin is retained within the endoplasmic reticulum rather than being secreted, where it functions as a molecular chaperone to protect against apoptosis by binding to misfolded proteins and preventing their aggregation . This intracellular protective role may represent an evolutionary adaptation to preserve cellular integrity during stress conditions, which, if sustained, eventually leads to resistin secretion and subsequent inflammatory signaling . Physiologically, this dual functionality suggests that resistin may serve as a molecular link between cellular stress and inflammation, where initial endoplasmic reticulum stress triggers intracellular resistin retention and chaperone activity, followed by eventual secretion that promotes inflammatory responses in neighboring cells and tissues . The surface hydrophobicity of resistin, particularly involving residues like Phe49, appears critical for its chaperone function but may also influence its inflammatory signaling capacity through altered protein-protein interactions . This mechanistic insight suggests therapeutic strategies might differentially target either the chaperone or inflammatory functions of resistin depending on the disease context, potentially preserving beneficial cellular protection while mitigating harmful inflammatory effects .
Distinguishing between the biological effects of different resistin oligomeric states requires specialized techniques that can isolate and characterize each form. Size exclusion chromatography combined with multi-angle light scattering (SEC-MALS) provides precise separation and molecular weight determination of monomeric, dimeric, and higher-order oligomeric resistin forms from biological samples or recombinant preparations . Native gel electrophoresis under non-reducing conditions can preserve the oligomeric states during separation, followed by Western blotting to visualize the distribution pattern of different resistin forms . Chemical cross-linking approaches using reagents like glutaraldehyde or BS3 can stabilize transient oligomeric forms for subsequent analysis by mass spectrometry to determine exact subunit composition and stoichiometry . Functional comparative studies should utilize purified resistin fractions of defined oligomeric states in parallel assays measuring inflammatory cytokine production, insulin signaling, and chaperone activity to establish structure-function relationships . Researchers can employ Resistin Mutant Human as a tool to study monomeric effects specifically, comparing its activity to wild-type resistin under identical experimental conditions, while site-directed mutagenesis targeting interface residues can generate variants with altered oligomerization properties for mechanistic studies . Additionally, fluorescently labeled resistin variants combined with fluorescence resonance energy transfer (FRET) techniques can monitor oligomerization dynamics in real-time within cellular environments, providing insights into how physiological or pathological conditions might shift the equilibrium between different resistin states .
Human resistin is composed of 108 amino acids as a prepeptide, with its hydrophobic signal peptide cleaved before secretion . The mature form of resistin circulates in the blood as a dimeric protein, consisting of two 92 amino acid polypeptides linked via disulfide bonds at Cys26 . Resistin has been implicated in linking obesity to insulin resistance, as it is found at elevated levels in the blood of obese individuals .
The Resistin Mutant (Human Recombinant) is a modified form of the natural resistin protein. This mutant has a specific cysteine residue mutated to prevent dimerization, which may allow it to act as an antagonist . The recombinant form of this protein is produced in Escherichia coli and consists of 93 amino acid residues, with a molecular weight of approximately 9.9 kDa .
Resistin plays a significant role in various physiological and pathological processes. In mice, resistin is produced and secreted by adipocytes and acts on skeletal muscle myocytes, hepatocytes, and adipocytes to reduce their sensitivity to insulin . In humans, resistin is primarily expressed in immune cells and is considered a pro-inflammatory molecule . It has been associated with chronic inflammatory diseases, metabolic disorders, infectious diseases, and certain cancers .
Research on resistin and its mutant forms continues to provide insights into its biological functions and potential therapeutic applications. The resistin mutant, due to its altered structure, may offer new avenues for studying the mechanisms of insulin resistance and inflammation . Additionally, understanding the role of resistin in host defense mechanisms could lead to the development of novel antimicrobial agents .