AKR1B1 Mouse

Aldose Reductase Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

AKR1B1 Mouse Recombinant produced in E.Coli is a single, non-glycosylated polypeptide chain containing 339 amino acids (1-316a.a.) and having a molecular mass of 38.1kDa.
AKR1B1 is fused to a 23 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
AKR1B1, a member of the aldo/keto reductase superfamily, plays a crucial role in glucose metabolism and is implicated in diabetic complications. It catalyzes the reduction of glucose to sorbitol, potentially contributing to the development of diabetic complications. This enzyme exhibits broad substrate specificity, reducing various carbonyl compounds. Studies using transgenic mice overexpressing human aldose reductase highlight AKR1B1's involvement in ischemic injury and its impact on post-ischemic recovery. Additionally, AKR1B1 is recognized as a crucial mediator in TNF-alpha signaling, influencing adhesion molecule expression and monocyte binding to the endothelium. Notably, it acts as a critical regulator of TNF-alpha-induced apoptosis in endothelial cells.
Description
Recombinant AKR1B1 Mouse protein, expressed in E. coli, is a single, non-glycosylated polypeptide chain. It consists of 339 amino acids (1-316a.a.) with a molecular weight of 38.1kDa. A 23 amino acid His-tag is fused to the N-terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The AKR1B1 protein solution is provided at a concentration of 1mg/ml. It is formulated in a buffer consisting of Phosphate buffered saline (pH7.4) and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To ensure optimal stability during long-term storage, the addition of a carrier protein such as HSA or BSA (0.1%) is advisable. It is important to minimize repeated freeze-thaw cycles.
Purity
The purity of the AKR1B1 protein is greater than 95% as determined by SDS-PAGE analysis.
Biological Activity
The specific activity of the enzyme is measured as greater than 500 pmol/min/ug. This is defined as the amount of enzyme required to catalyze the reduction of 1.0 pmole of DL-glyceraldehyde per minute in the presence of NADPH at a pH of 7.0 and a temperature of 37°C.
Synonyms
Aldose reductase, AKR1B1, AR, Aldehyde reductase, Akr1b3, Aldor1, Aldr1, Akr1b1, Ahr-1, Ahr1, ALR2.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSMASHLEL NNGTKMPTLG LGTWKSPPGQ VTEAVKVAID LGYRHIDCAQ VYQNEKEVGV ALQEKLKEQV VKRQDLFIVS KLWCTFHDKS MVKGAFQKTL SDLQLDYLDL YLIHWPTGFK PGPDYFPLDA SGNVIPSDTD FVDTWTAMEQ LVDEGLVKTI GVSNFNPLQI ERILNKPGLK YKPAVNQIEC HPYLTQEKLI EYCHSKGIVV TAYSPLGSPD RPWAKPEDPS LLEDPRIKAI AAKYNKTTAQ VLIRFPIQRN LVVIPKSVTP VRIAENLKVF DFEVSSEDMA TLLSYNRNWR VCALMSCAKH KDYPFHAEV.

Q&A

What is AKR1B1 and how does it differ between humans and mice?

AKR1B1 (Aldo-keto reductase family 1 member B1), commonly known as aldose reductase, is a monomeric NADPH-dependent cytosolic enzyme that catalyzes the reduction of various aldehydes and ketones . In mice, the orthologous gene is referred to as Akr1b1, though the protein functions are largely conserved. The mouse Akr1b1 protein consists of 316 amino acids with a molecular weight of approximately 38.1 kDa . The primary functional difference lies in tissue-specific expression patterns and substrate specificities, which researchers must consider when translating findings between species.

What are the main physiological functions of AKR1B1 in mice?

AKR1B1 primarily functions in the polyol pathway, catalyzing the rate-limiting reduction of glucose to sorbitol using NADPH as a cofactor. This enzymatic activity leads to a decrease in the NADPH/NADP+ ratio, potentially causing oxidative stress through increased reactive oxygen species (ROS) production . In normal physiology, AKR1B1 participates in detoxification of reactive aldehydes, osmoregulation, and certain aspects of lipid metabolism. Its activity in converting glucose to sorbitol becomes particularly relevant under hyperglycemic conditions, contributing to diabetic complications in various tissues including the lens, kidney, and nervous system .

How is AKR1B1 expression regulated in mouse tissues?

AKR1B1 expression in mice exhibits tissue-specific regulation patterns. Transcription factors like Twist2 have been shown to induce AKR1B1 expression, particularly in certain cancer contexts . The expression can be further amplified through positive feedback loops involving NF-κB activation. AKR1B1 expression also responds to oxidative stress conditions, inflammatory stimuli, and hyperglycemia. Different mouse tissues show variable basal expression levels, with particularly notable expression in lens, kidney, and certain transformed cell types. Research suggests that epigenetic modifications and microRNA regulation may also play roles in controlling tissue-specific expression patterns.

What are the most effective methods for measuring AKR1B1 activity in mouse tissue samples?

For quantifying AKR1B1 enzyme activity in mouse tissue samples, researchers should employ a combined approach:

  • Spectrophotometric enzyme assay: Measure specific activity by monitoring NADPH consumption (decrease in absorbance at 340 nm) during the reduction of substrates like DL-glyceraldehyde. Standardized conditions include pH 7.0 at 37°C with activity expressed as pmol/min/μg protein .

  • ELISA-based quantification: Commercial ELISA kits provide sensitivity down to 9.375 pg/ml with detection ranges of 15.625-1000 pg/ml for mouse AKR1B1 in serum, plasma, and tissue homogenates .

  • Western blotting: For relative protein expression comparison, use tissue-specific extraction protocols optimized for cytosolic proteins with PVDF membranes and specific anti-AKR1B1 antibodies.

  • RT-qPCR: Quantify gene expression using RNA extraction with A260/280 ratios between 1.8-2.0, followed by reverse transcription and qPCR with Akr1b1-specific primers .

For enhanced reliability, researchers should include both activity assays and protein/gene expression measurements, as post-translational modifications may affect enzyme activity independently of expression levels.

What transgenic mouse models are available for studying AKR1B1 function?

Several transgenic mouse models have been developed to study AKR1B1 function:

  • Human AKR1B1 overexpression models: Transgenic mice (e.g., PAR37 and PAR39 strains) expressing high levels of human AKR1B1 have been created, which develop phenotypes including lens opacity and aberrant cellular differentiation . These models are particularly valuable for studying AKR1B1's role in diabetic complications.

  • Tissue-specific AKR1B1 expression models: Mice with targeted expression in specific tissues using tissue-specific promoters allow for examining organ-specific functions and pathologies.

  • AKR1B1 knockout models: These provide essential insights into the physiological roles of the enzyme and serve as negative controls for pharmacological inhibition studies.

  • Inducible expression systems: Models utilizing tetracycline-responsive or Cre-loxP systems enable temporal control of AKR1B1 expression, facilitating developmental studies and avoiding compensatory adaptations during development.

When selecting a model, researchers should consider expression levels, as phenotypic effects may depend on the degree of expression. For instance, PAR40 mice with lower AKR1B1 expression exhibit normal lens morphology, while higher-expressing PAR37 and PAR39 strains develop lens abnormalities .

What is the optimal protocol for isolating and purifying active AKR1B1 protein from mouse tissues?

The optimal protocol for isolating functional AKR1B1 from mouse tissues involves:

  • Tissue preparation: Flash-freeze fresh tissue in liquid nitrogen and pulverize using a pre-chilled mortar and pestle.

  • Homogenization: Homogenize tissue in phosphate-buffered saline (pH 7.4) containing 10% glycerol and protease inhibitors . Maintain cold temperature (4°C) throughout extraction.

  • Differential centrifugation: Clear cellular debris with low-speed centrifugation (3,000g, 15 min), followed by high-speed centrifugation (100,000g, 60 min) to obtain cytosolic fraction.

  • Affinity chromatography: For highest purity, utilize His-tagged recombinant constructs with nickel affinity columns , or employ AKR1B1-specific antibody affinity purification.

  • Ion exchange chromatography: Further purify using DEAE or Q-Sepharose columns with gradient elution.

  • Storage: Store with 10% glycerol at -80°C in aliquots to minimize freeze-thaw cycles .

Verification of purity should include SDS-PAGE (>95% purity), Western blotting, and specific activity assays to confirm biological function. MALDI-TOF can confirm molecular weight (approximately 38.1 kDa for mouse AKR1B1) .

How does AKR1B1 contribute to diabetic complications in mouse models?

AKR1B1 contributes to diabetic complications in mouse models through several interconnected mechanisms:

  • Polyol pathway activation: Under hyperglycemic conditions, AKR1B1 catalyzes glucose conversion to sorbitol, which accumulates intracellularly causing osmotic stress and cellular damage.

  • Redox imbalance: The reaction consumes NADPH, decreasing the NADPH/NADP+ ratio and depleting cellular antioxidant capacity. This leads to increased oxidative stress through elevated reactive oxygen species (ROS) levels, as demonstrated in studies showing that AKR1B1 knockdown significantly decreases ROS levels in susceptible cells .

  • NF-κB activation: AKR1B1-mediated increase in ROS activates the redox-sensitive NF-κB pathway, triggering inflammatory responses. Additionally, AKR1B1 can activate NF-κB signaling via PGF2α-mediated pathways .

  • Tissue-specific pathology: In transgenic mice expressing high levels of human AKR1B1, lens abnormalities develop, including opacity, aberrant nucleated cells beneath the anterior epithelium, and large vacuoles in the lens bow region . These pathologies can be suppressed by treatment with aldose reductase inhibitors like sorbinil .

  • Epithelial-mesenchymal transition (EMT): AKR1B1 overexpression promotes EMT through positive feedback loops involving Twist2 and NF-κB, contributing to cellular dysfunction in affected tissues .

These pathological mechanisms highlight why AKR1B1 inhibition represents a potential therapeutic approach for diabetic complications.

What is the role of AKR1B1 in inflammatory responses in mouse models?

AKR1B1 plays a significant role in modulating inflammatory responses in mouse models through multiple mechanisms:

  • NF-κB pathway activation: AKR1B1 activity promotes NF-κB signaling through both redox-dependent and independent mechanisms, leading to increased production of pro-inflammatory cytokines. Studies in sepsis-associated acute kidney injury (SA-AKI) models show that inhibiting AKR1B1 reduces levels of IL-1β, IL-6, and TNF-α in serum and kidney tissues .

  • Oxidative stress amplification: By consuming NADPH, AKR1B1 reduces cellular antioxidant capacity, increasing ROS production that further amplifies inflammatory signaling pathways.

  • Protein Kinase C (PKC) activation: AKR1B1 activity influences PKC signaling, which intersects with inflammatory pathways. Inhibition of AKR1B1 affects PKC/NF-κB pathway protein expression in inflammatory models .

  • Prostaglandin synthesis: AKR1B1 participates in prostaglandin metabolism, particularly PGF2α, which modulates inflammatory responses in various tissues .

In cecum ligation puncture (CLP) models of sepsis, AKR1B1 inhibition with agents like epalrestat has been shown to reduce inflammatory responses, demonstrating therapeutic potential for inflammatory conditions . Treatment protocols delivering AKR1B1 inhibitors either preventively (pre-ARI) or therapeutically (post-ARI) have shown efficacy in reducing inflammatory markers and tissue damage in experimental models.

How does AKR1B1 influence cancer progression in mouse models?

AKR1B1 promotes cancer progression in mouse models through several coordinated mechanisms:

  • Epithelial-mesenchymal transition (EMT) activation: AKR1B1 operates within a positive feedback loop where Twist2 transcriptionally induces AKR1B1 expression, leading to NF-κB activation, which in turn up-regulates Twist2. This feedback loop activates the EMT program, characterized by loss of epithelial markers (E-cadherin) and gain of mesenchymal markers (vimentin, N-cadherin) .

  • Cancer stem cell (CSC) properties enhancement: The AKR1B1-mediated EMT program promotes cancer stem cell-like properties, increasing tumorigenicity and metastatic potential. This is particularly evident in basal-like breast cancer (BLBC) models, where AKR1B1 overexpression correlates with poor prognosis .

  • Redox modulation: AKR1B1 activity decreases the NADPH/NADP+ ratio, causing oxidative stress through increased ROS levels. This redox imbalance activates signaling pathways that favor tumor progression .

  • Inflammatory microenvironment: By activating NF-κB signaling, AKR1B1 promotes pro-inflammatory cytokine production, creating a tumor-supportive microenvironment.

  • Metabolic adaptation: AKR1B1 participates in lipid metabolism through PGF2α-mediated pathways, potentially contributing to metabolic reprogramming in cancer cells .

Experimental evidence shows that AKR1B1 expression promotes, while its knockdown inhibits, tumorigenicity and metastasis in mouse models. Notably, AKR1B1 protein levels are consistently elevated in basal-like breast cancer cell lines but absent in luminal cell lines, suggesting subtype-specific roles in breast cancer progression .

How can researchers effectively use aldose reductase inhibitors (ARIs) in mouse models?

Researchers can optimize aldose reductase inhibitor (ARI) studies in mouse models through these methodological considerations:

  • Inhibitor selection: Choose ARIs based on specificity, potency, and pharmacokinetic properties. Common ARIs include:

    • Sorbinil: Effective in suppressing lens phenotypes in AKR1B1 transgenic mice

    • Epalrestat: Commonly used at 100 mg/kg/d in rodent models, dissolved in 0.5% sodium carboxymethyl cellulose

    • Fidarestat, Zopolrestat, and Ranirestat: Alternative options with varying specificity profiles

  • Administration protocols:

    • Preventive (pre-ARI): Administer before disease induction to assess prophylactic potential

    • Therapeutic (post-ARI): Administer after disease onset to assess treatment efficacy

    • Chronic administration: For models of chronic diseases like diabetes complications

    • Administration routes: Oral gavage, drinking water incorporation, or intraperitoneal injection depending on study design

  • Dosing optimization: Perform dose-response studies (25-200 mg/kg/d) to establish effective dose range while monitoring for potential off-target effects.

  • Validation of target engagement:

    • Measure AKR1B1 activity in target tissues after inhibitor administration

    • Assess downstream markers (e.g., sorbitol levels, NADPH/NADP+ ratio)

    • Monitor ROS levels and NF-κB activation status

  • Experimental controls:

    • Vehicle controls (e.g., 0.5% Na-CMC solution)

    • Genetic models (AKR1B1 knockouts) as positive controls for ARI effects

    • Include both wildtype and AKR1B1-overexpressing mice for comparative analysis

Recent studies demonstrate successful application of epalrestat in cecum ligation perforation (CLP) models of sepsis-associated acute kidney injury, with significant improvements in kidney function markers and inflammatory parameters .

What are the key considerations for investigating AKR1B1 in specific tissue microenvironments?

Investigating AKR1B1 in specific tissue microenvironments requires attention to several critical factors:

  • Tissue-specific expression patterns:

    • Baseline expression varies significantly across tissues

    • BLBC cell lines show high AKR1B1 expression, while luminal breast cancer lines show minimal expression

    • Lens, kidney, and nervous tissues have distinct expression profiles relevant to diabetic complications

  • Microenvironmental factors affecting AKR1B1 function:

    • Glucose concentration: Hyperglycemic conditions enhance AKR1B1 relevance

    • Oxygen tension: Hypoxic environments may alter AKR1B1 activity and significance

    • Inflammatory mediators: Cytokines may induce AKR1B1 expression

    • Extracellular matrix composition: May influence cellular responses to AKR1B1 activity

  • Single-cell resolution techniques:

    • Single-cell RNA sequencing to identify cell populations with differential AKR1B1 expression

    • Immunofluorescence microscopy with co-staining for cell-type specific markers

    • Laser capture microdissection for region-specific analysis

  • Cell-cell interaction considerations:

    • Paracrine effects of AKR1B1-expressing cells on neighboring cells

    • Co-culture systems to model heterotypic interactions

    • Spatial transcriptomics to map AKR1B1 expression patterns relative to tissue architecture

  • Tissue-specific manifestations:

    • Lens: Evaluate anterior epithelial plaques, opacity, and vacuole formation

    • Kidney: Assess tubular damage, BUN/creatinine levels, and inflammatory infiltrates

    • Cancer: Examine EMT markers, stem cell properties, and invasive capacity

A comprehensive approach combines tissue-specific pathological scoring systems (e.g., H&E staining with damage scales for kidney) with molecular profiling (proteomics, transcriptomics) and functional assays relevant to the tissue of interest.

How do post-translational modifications affect AKR1B1 function in mouse models?

Post-translational modifications (PTMs) significantly impact AKR1B1 function in mouse models through multiple mechanisms:

  • Phosphorylation:

    • Serine/threonine phosphorylation can alter catalytic efficiency and substrate specificity

    • Protein kinase C (PKC) activation, which intersects with AKR1B1 signaling pathways, may regulate AKR1B1 through phosphorylation events

    • Phosphorylation status should be assessed using phospho-specific antibodies or phosphoproteomic approaches

  • Oxidative modifications:

    • As a redox-sensitive enzyme, cysteine residues in AKR1B1 can undergo S-glutathionylation, S-nitrosylation, or oxidation

    • These modifications typically reduce enzyme activity and may serve as negative feedback during oxidative stress

    • Assessment requires specialized techniques such as redox proteomics or biotin-switch assays

  • Glycosylation:

    • N-linked glycosylation affects protein stability and potentially subcellular localization

    • Methods for detection include glycoprotein staining, lectin affinity, or mass spectrometry following PNGase F treatment

  • Ubiquitination and SUMOylation:

    • These modifications regulate protein turnover and subcellular localization

    • Proteasome inhibitors can be used to accumulate ubiquitinated forms for detection

  • Technical approaches for studying PTMs:

    • Mass spectrometry-based proteomics for global PTM profiling

    • Site-directed mutagenesis of modification sites to create PTM-deficient variants

    • In vitro modification assays to establish functional consequences

    • Subcellular fractionation to determine if PTMs affect localization

When characterizing recombinant AKR1B1 from expression systems, researchers should verify that the purified protein (>95% by SDS-PAGE) maintains proper folding and modifications that reflect the in vivo state . Molecular weight confirmation by methods such as MALDI-TOF can identify the presence of major PTMs that significantly alter protein mass.

How can researchers address variability in AKR1B1 expression across different mouse strains?

Researchers can address strain-specific variability in AKR1B1 expression through these methodological approaches:

  • Comprehensive strain characterization:

    • Perform baseline profiling of AKR1B1 expression and activity across common laboratory strains (C57BL/6, BALB/c, 129, FVB, etc.)

    • Create a reference database of strain-specific expression patterns in key tissues

    • Consider both protein levels (Western blotting, ELISA) and enzymatic activity measurements

  • Experimental design strategies:

    • Always use littermate controls within the same strain

    • When comparing across strains, normalize data to strain-specific baselines

    • Consider backcrossing transgenic lines to multiple genetic backgrounds to assess phenotype robustness

    • For transgenic models, validate expression levels as phenotypic effects may be expression-dependent (as seen with PAR37/PAR39 vs. PAR40 strains)

  • Statistical approaches:

    • Implement mixed-effects models that account for strain as a random variable

    • Perform power analyses specific to each strain based on observed variability

    • Consider stratified analysis when combining data from multiple strains

  • Molecular strategies:

    • Use CRISPR/Cas9 to generate equivalent mutations or modifications across different strains

    • Employ strain-specific reference genes for accurate RT-qPCR normalization

    • Use tissue-specific promoters to achieve comparable expression levels across strains

  • Reporting standards:

    • Explicitly state strain background, including substrain designations

    • Report generation number for transgenic lines

    • Document housing conditions, as environmental factors may interact with strain backgrounds

These approaches ensure robust, reproducible findings while acknowledging the biological reality of strain differences, improving translational relevance of AKR1B1 research.

What are the best practices for comparing in vitro and in vivo findings in AKR1B1 research?

Best practices for reconciling in vitro and in vivo AKR1B1 research include:

  • Matching experimental conditions:

    • Use physiologically relevant concentrations of substrates, cofactors, and inhibitors

    • For in vitro enzyme assays, perform activity measurements at physiological pH (7.0-7.4) and temperature (37°C)

    • When possible, use primary cells rather than immortalized lines for in vitro studies

    • Consider 3D culture systems that better recapitulate tissue architecture

  • Pathway validation across systems:

    • Verify that key molecular mechanisms (e.g., AKR1B1-mediated NF-κB activation) operate similarly in both contexts

    • Use identical readouts and biomarkers across in vitro and in vivo experiments

    • Confirm that pharmacological inhibitors demonstrate comparable target engagement in both settings

  • Translational approaches:

    • Employ ex vivo tissue explants as intermediate models

    • Use conditional or inducible systems that can be manipulated at specific timepoints in both contexts

    • Develop organoid models from primary tissues to bridge the complexity gap

  • Quantitative considerations:

    • Account for differences in relative AKR1B1 expression between cultured cells and tissues

    • Adjust for pharmacokinetic factors when translating inhibitor studies

    • Normalize enzyme activity data to account for different tissue/cellular contexts

  • Documentation and reporting:

    • Explicitly acknowledge limitations when extrapolating between systems

    • Report detailed methodological conditions for both in vitro (e.g., passage number, confluence) and in vivo (e.g., age, sex, housing) experiments

    • Include positive controls that demonstrate known AKR1B1 behaviors in each system

Following these practices enhances the translational value of combined in vitro and in vivo approaches, as exemplified in studies examining AKR1B1's role in cancer progression where cellular findings regarding EMT were validated in mouse models .

How should researchers interpret contradictory findings about AKR1B1 function across different experimental models?

When faced with contradictory findings about AKR1B1 function across experimental models, researchers should implement this systematic analytical framework:

  • Context-dependent assessment:

    • Recognize that AKR1B1 functions may legitimately differ across tissues, developmental stages, or disease states

    • Examine whether apparently contradictory findings occur in different biological contexts

    • Consider that AKR1B1 may have dual roles depending on expression level, as seen in transgenic models where phenotypic effects correlate with expression intensity

  • Methodological reconciliation:

    • Scrutinize differences in experimental methods, including:

      • Protein source (recombinant vs. endogenous)

      • Inhibitor specificity and dosing regimens

      • Timing of interventions (preventive vs. therapeutic approaches)

      • Outcome measures and their sensitivity

    • Replicate key experiments using standardized protocols across models

  • Molecular mechanism dissection:

    • Determine if contradictions reflect different aspects of AKR1B1's multiple functions

      • Enzymatic activity (polyol pathway)

      • Signaling functions (NF-κB activation)

      • Transcriptional regulatory effects

    • Use domain-specific mutations or inhibitors to separate distinct functions

  • Integrative data analysis:

    • Perform meta-analyses across studies with careful weighting for methodological quality

    • Use systems biology approaches to place contradictory findings within broader pathway contexts

    • Consider mathematical modeling to identify conditions under which different outcomes might emerge

  • Targeted validation experiments:

    • Design experiments specifically to test contradictory findings head-to-head

    • Include genetic approaches (e.g., CRISPR/Cas9) alongside pharmacological ones

    • Utilize rescue experiments to confirm specificity of observed effects

This framework enables researchers to distinguish true biological complexity from methodological artifacts, ultimately leading to a more nuanced understanding of AKR1B1's multifaceted roles in health and disease.

What emerging technologies can advance AKR1B1 research in mouse models?

Several cutting-edge technologies promise to revolutionize AKR1B1 research in mouse models:

  • CRISPR-based approaches:

    • Base editing for introducing precise point mutations in AKR1B1 regulatory regions

    • CRISPRa/CRISPRi systems for temporal and cell-type specific modulation of expression

    • Prime editing for creating specific disease-associated variants

    • CRISPR screens to identify novel interactors and regulators of AKR1B1 function

  • Advanced imaging techniques:

    • Intravital microscopy with fluorescently tagged AKR1B1 to track dynamics in living tissues

    • FRET-based biosensors to monitor AKR1B1 activity in real-time

    • Correlative light and electron microscopy (CLEM) for subcellular localization studies

    • Label-free imaging methods to detect metabolic changes associated with AKR1B1 activity

  • Single-cell technologies:

    • Single-cell RNA sequencing to identify cell populations with differential AKR1B1 expression

    • Single-cell proteomics to correlate AKR1B1 protein levels with other cellular factors

    • Spatial transcriptomics to map expression patterns within complex tissues

    • Single-cell metabolomics to detect AKR1B1-mediated metabolic alterations

  • Biomolecular condensate analysis:

    • Investigation of AKR1B1's potential role in phase separation phenomena

    • Examination of how cellular stress affects AKR1B1 compartmentalization

    • Development of optogenetic tools to control AKR1B1 localization

  • Integrated multi-omics approaches:

    • Combined proteomics, transcriptomics, and metabolomics to create comprehensive pathway maps

    • Systems biology modeling of AKR1B1's role in complex networks

    • Machine learning applications to predict AKR1B1-dependent phenotypes from multi-omics data

These technologies will facilitate more precise understanding of AKR1B1's tissue-specific functions and disease contributions, potentially leading to more targeted therapeutic strategies.

What are the most promising therapeutic applications of AKR1B1 research in mouse models?

Mouse model research has identified several promising therapeutic applications targeting AKR1B1:

  • Diabetic complications:

    • Targeted inhibition with second and third-generation aldose reductase inhibitors (ARIs)

    • Tissue-specific delivery systems to reduce systemic side effects

    • Combination therapies addressing both AKR1B1 activity and downstream consequences

    • Preventive approaches utilizing the pre-ARI strategy demonstrated in experimental models

  • Inflammatory conditions:

    • AKR1B1 inhibition for sepsis-associated acute kidney injury, as demonstrated in CLP models showing significant improvement in kidney function and reduction in inflammatory markers

    • Targeted approaches for inflammatory diseases where ROS and NF-κB signaling play central roles

    • Development of dual-action compounds that inhibit both AKR1B1 and inflammatory mediators

  • Cancer therapeutics:

    • Targeting the AKR1B1-Twist2-NF-κB feedback loop in basal-like breast cancer

    • Combination therapies targeting both AKR1B1 and cancer stem cell properties

    • Biomarker-guided approaches using AKR1B1 expression to identify responsive tumors

    • Development of inhibitors that specifically disrupt AKR1B1's role in EMT without affecting other functions

  • Precision medicine approaches:

    • Patient stratification based on AKR1B1 expression profiles

    • Development of companion diagnostics to predict response to AKR1B1 inhibitors

    • Genotype-specific interventions targeting particular AKR1B1 variants

  • Novel delivery technologies:

    • Nanoparticle-based delivery of AKR1B1 inhibitors to specific tissues

    • mRNA-based approaches to modulate AKR1B1 expression

    • Extracellular vesicle delivery systems targeting cells with high AKR1B1 expression

Translational research utilizing mouse models has established proof-of-concept for these approaches, with particularly strong evidence for applications in diabetic complications, inflammatory conditions, and specific cancer subtypes with AKR1B1 overexpression .

How can multi-omics approaches enhance our understanding of AKR1B1 biology in mouse models?

Multi-omics approaches can revolutionize AKR1B1 research through integrative methodologies:

  • Comprehensive pathway mapping:

    • Proteomics to identify AKR1B1 interaction networks and post-translational modifications

    • Transcriptomics to elucidate expression patterns and regulatory mechanisms

    • Metabolomics to characterize substrate profiles and downstream metabolic effects

    • Integration of these datasets to create comprehensive AKR1B1 functional maps

  • Disease mechanism elucidation:

    • Label-free LC-MS/MS proteomics to identify differentially expressed proteins in disease models, as successfully applied in sepsis-associated acute kidney injury models

    • Temporal multi-omics analyses to track disease progression and intervention effects

    • Single-cell multi-omics to identify cell populations particularly dependent on AKR1B1 function

  • Methodological approaches:

    • Tissue-specific analyses comparing affected and unaffected tissues

    • Temporal sampling before and after interventions with AKR1B1 inhibitors

    • Comparison of genetic models (knockouts, transgenics) with pharmacological interventions

    • Cross-species comparative analyses to identify conserved mechanisms

  • Data integration strategies:

    • Network analysis algorithms to identify key nodes and regulatory hubs

    • Machine learning approaches to predict phenotypic outcomes from multi-omics signatures

    • Pathway enrichment analyses to place AKR1B1 within broader biological contexts

    • Development of computational models that predict systemic effects of AKR1B1 modulation

  • Translational applications:

    • Identification of novel biomarkers for monitoring AKR1B1-related pathologies

    • Discovery of previously unrecognized therapeutic targets within AKR1B1 pathways

    • Development of predictive models for patient stratification in clinical trials

Product Science Overview

Introduction

Aldose reductase (AR) is a cytosolic monomeric enzyme that belongs to the aldo-keto reductase (AKR) superfamily. This superfamily includes more than 150 NAD(P)(H)-dependent oxidoreductases found in all prokaryotic and eukaryotic kingdoms, including yeast, plants, invertebrates, and vertebrates . The enzyme is known for its role in the polyol pathway, where it catalyzes the reduction of glucose to sorbitol .

Structure and Function

Aldose reductase is a reduced nicotinamide-adenine dinucleotide phosphate (NADPH)-dependent enzyme that catalyzes the reduction of various aldehydes and ketones to their corresponding alcohols . The enzyme consists of 316 amino acid residues and weighs approximately 35,853 Da . The active site pocket of aldose reductase is relatively hydrophobic, lined by several aromatic and non-polar residues .

Role in Diabetic Complications

Aldose reductase has been extensively studied due to its involvement in diabetic complications . Under diabetic conditions, the enzyme converts excess glucose into sorbitol, which is then converted to fructose . This accumulation of sorbitol and fructose can lead to osmotic stress and contribute to various diabetic complications, such as diabetic retinopathy and nephropathy .

Tissue Distribution and Expression

Aldose reductase is ubiquitously expressed in various human organs, including the kidney, lens, retina, nerve, heart, placenta, brain, skeletal muscle, testis, blood vessels, lung, and liver . However, AR-like proteins exhibit tissue-specific patterns of expression . For instance, certain isoforms are enriched in the adrenal gland, enterohepatic, and adipose tissues .

Physiological Functions Beyond Detoxification

Beyond its role in detoxification, recent studies suggest that aldose reductase and its isoforms may have additional physiological functions . These enzymes are capable of modifying or generating signaling molecules, shifting their status from mere scavengers to important messengers . This has implications for their roles in glucido-lipidic metabolism and adipose tissue homeostasis .

Clinical Significance

Aldose reductase is involved in many oxidative stress diseases, cell signal transduction, and cell proliferation processes, including cardiovascular disorders, sepsis, and cancer . Inhibition of aldose reductase has been suggested as a therapeutic strategy to reduce inflammation associated with the activation of retinal microglia .

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.