GLO1 Human

Glyoxalase-I Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Genetic Features

GLO1 is encoded by the GLO1 gene located at chromosome 6p21.2. The enzyme comprises 184 amino acids, forming a 21-kDa homodimeric Zn²⁺-dependent metalloprotein . Key structural elements include:

FeatureDetail
Active site ligandsHis127, Glu173, Gln34, and Glu100 coordinate Zn²⁺ at the dimer interface .
Promoter elementsAntioxidant-response element (ARE), insulin-response element (IRE) .
Amino acid sequence (partial)MAEPQPPSGG...MKGLAFIQDPDGYWIEILNPNKMATLM (184 residues total) .

Biological Functions

GLO1 catalyzes the first step of the glyoxalase cycle:

  1. Substrate binding: MGO spontaneously reacts with glutathione (GSH) to form hemithioacetal.

  2. Isomerization: GLO1 converts hemithioacetal into S-D-lactoylglutathione .

  3. Hydrolysis: Glyoxalase II (GLO2) processes this intermediate into D-lactate, regenerating GSH .

This system reduces intracellular MGO levels by ~99%, preventing MGO-induced protein glycation, oxidative stress, and apoptosis .

Cancer

  • Overexpression: Elevated GLO1 activity is observed in pancreatic, breast, and prostate cancers, enabling tumor survival by mitigating MGO cytotoxicity .

  • Chemoresistance: GLO1 upregulation in leukemia and melanoma cells confers resistance to etoposide and adriamycin .

Diabetes and Vascular Complications

  • Hyperglycemia: High glucose increases MGO production, overwhelming GLO1 capacity and leading to advanced glycation end-products (AGEs) .

  • Endothelial dysfunction: GLO1 knockdown in aortic endothelial cells increases MGO, inflammation (↑ ICAM-1, VCAM-1), and apoptosis .

Neurological Disorders

  • Autism and schizophrenia: The GLO1 SNP rs2736654 (Ala111Glu) is linked to autism susceptibility .

  • Pain and anxiety: Murine studies show GLO1 modulates neuropathic pain and anxiety-like behavior via MGO regulation .

Genetic Polymorphisms

Key GLO1 variants and their clinical implications:

PolymorphismLocationAssociationReference
rs2736654 (-419C>A)Exon 4 (Ala111Glu)Autism, multiple sclerosis, cerebral malformations .
C332CExon 4Diabetic neuropathy in type 2 diabetes .
C-7TPromoterAtherosclerosis in type 2 diabetics .

Therapeutic Implications

  • Inhibition strategies: Small-molecule GLO1 inhibitors (e.g., S-p-bromobenzylglutathione) are under investigation for cancer therapy .

  • Gene editing: GLO1 knockout in stem cells exacerbates carbonyl stress, impairing mitochondrial function and neurite outgrowth .

Research Challenges

  • Conflicting evidence: While GLO1 overexpression aids tumor survival, its role in diabetic complications remains debated .

  • Human studies: Limited replication of behavioral associations (e.g., anxiety) in human trials compared to animal models .

Product Specs

Introduction
GLO1, an enzyme encoded by the GLO1 gene located on chromosome 6p21.3-p21.1, catalyzes the detoxification of methylglyoxal (MG) into S-lactoyl-glutathione. This enzymatic reaction is crucial for cellular protection against MG-induced damage. GLO1 is implicated in various biological processes, including cellular detoxification, inflammation, and the pathogenesis of diseases like autism, mood disorders, and cancer. Its upregulation in tumor cells highlights its potential as a therapeutic target.
Description
Recombinant human Glyoxalase-I, expressed in E. coli, is a non-glycosylated polypeptide chain comprising 184 amino acids with a molecular weight of 20.7 kDa. The protein is purified to a high degree using proprietary chromatographic methods.
Physical Appearance
The product is a clear, colorless, and sterile-filtered solution.
Formulation
Glyoxalase-1 is supplied in a solution buffered with 20mM Tris-HCl (pH 8.0), 1mM DTT (reducing agent), and 10% glycerol (cryoprotectant).
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, freezing at -20°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is advised for long-term storage to maintain protein stability. Repeated freeze-thaw cycles should be avoided to prevent protein degradation.
Purity
The purity of Glyoxalase-1 is determined to be greater than 90% using SDS-PAGE analysis.
Synonyms
GLYI, GLOD1, GLO1, Glyoxalase-1, Lactoylglutathione lyase, Methylglyoxalase, Aldoketomutase, Ketone-aldehyde mutase, Glyoxalase I, S-D-lactoylglutathione methylglyoxal lyase, Glx I.
Source
Escherichia Coli.
Amino Acid Sequence
MAEPQPPSGG LTDEAALSCC SDADPSTKDF LLQQTMLRVK DPKKSLDFYT RVLGMTLIQK CDFPIMKFSL YFLAYEDKND IPKEKDEKIAWALSRKATLE LTHNWGTEDD ETQSYHNGNS DPRGFGHIGI AVPDVYSACK RFEELGVKFV KKPDDGKMKG LAFIQDPDGY WIEILNPNKM ATLM.

Q&A

What is human GLO1 and what is its primary function in cellular metabolism?

Glyoxalase 1 (GLO1) is a zinc metalloenzyme essential for cellular detoxification of methylglyoxal (MG), a reactive dicarbonyl compound produced as a byproduct of glycolysis. As the first enzyme in the glyoxalase system, GLO1 catalyzes the conversion of the hemithioacetal formed from MG and glutathione (GSH) to S-D-lactoylglutathione . This enzymatic action represents a critical defense mechanism against dicarbonyl stress and prevents the formation of advanced glycation end products (AGEs). The detoxification process is completed by Glyoxalase 2 (GLO2), which converts S-D-lactoylglutathione to D-lactic acid, regenerating GSH in the process . GLO1 plays a vital role in preventing protein and nucleotide modifications that can lead to cellular dysfunction and apoptosis .

How does GLO1 expression vary across human tissues and what implications does this have for research design?

GLO1 shows differential expression and localization patterns across human tissues. In skin, for example, GLO1 enzymes are differentially localized in the epidermis . When designing studies investigating GLO1, researchers should account for tissue-specific expression levels and functions. This requires careful selection of appropriate control tissues and consideration of tissue-specific regulation mechanisms.

For skin-based studies, researchers should isolate and analyze distinct skin layers separately rather than using whole skin homogenates. In comparative studies across multiple tissues, normalization strategies must account for baseline differences in GLO1 expression. Additionally, when using cell culture models, researchers should verify that GLO1 expression in their model accurately reflects the in vivo situation in the tissue of interest .

What are the most reliable biomarkers for assessing GLO1 activity in human samples?

When assessing GLO1 activity in human samples, researchers should employ multiple complementary approaches:

  • Direct enzyme activity assays measuring the rate of S-D-lactoylglutathione formation

  • Quantification of MG levels using liquid chromatography-mass spectrometry

  • Detection of MG-derived protein adducts (particularly arginine-directed modifications)

  • Measurement of D-lactate as the end product of the glyoxalase pathway

For cellular studies, researchers can use siRNA-mediated knockdown of GLO1 followed by determination of MG concentrations in both cells and culture medium . Western blotting to detect GLO1 protein levels and quantitative PCR to measure GLO1 mRNA expression provide additional validation . For a comprehensive assessment, researchers should examine downstream effects on protein glycation and functional outcomes specific to the tissue under investigation.

How does GLO1 dysfunction contribute to endothelial damage in diabetic complications?

GLO1 dysfunction creates a cascade of molecular events leading to endothelial damage in diabetic settings. Studies of human aortic endothelial cells (HAECs) with GLO1-knockdown show that reduced GLO1 activity increases methylglyoxal (MG) concentration in both cells and culture medium . This MG accumulation triggers multiple pathological processes:

  • Differential abundance of cytoskeleton stabilization proteins and intermediate filaments

  • Altered posttranslational modification of collagen and increased fibrillar collagens 1 and 5

  • Elevated extracellular concentration of endothelin-1, promoting vasoconstriction

  • Increased expression of inflammatory markers (ICAM-1, VCAM-1, MCP-1)

  • Enhanced apoptotic signaling in endothelial cells

These changes collectively manifest as endothelial dysfunction, inflammation, and accelerated vascular damage. Methodologically, researchers investigating these mechanisms should employ both targeted approaches (examining specific inflammatory markers) and unbiased proteomic screening to identify novel GLO1-dependent pathways in endothelial cells .

What is the evidence supporting GLO1 as a therapeutic target in cancer research?

GLO1 has emerged as a promising anticancer target due to its differential expression and functional importance in malignant cells. Research shows abnormal expression or higher GLO1 activity in multiple human cancers including lung, breast, prostate, pancreatic, gastric, colon, and skin cancers, as well as in melanoma and drug-resistant human leukemia cells .

The rationale for targeting GLO1 in cancer is multifaceted:

  • Cancer cells exhibit increased glycolysis (Warburg effect), leading to higher MG production

  • MG has been shown to modify DNA, RNA, and proteins, and induce apoptosis in tumor cells

  • Cancer cells appear dependent on GLO1 to manage elevated MG levels

  • GLO1 inhibitors can induce apoptosis by promoting MG accumulation selectively in cancer cells

Recent structural studies of human GLO1 in complex with inhibitors like TLSC702 provide a molecular foundation for rational drug design. These structures reveal that inhibitors coordinate with the Zn²⁺ in the active site through carboxyl oxygen atoms while establishing van der Waals interactions with hydrophobic residues . Researchers should incorporate these structural insights when designing novel GLO1-targeting compounds and validate their effects on MG accumulation and cancer cell viability.

How does age-related decline in GLO1 function contribute to tissue aging and age-related diseases?

Age-related decline in GLO1 function represents a significant contributor to tissue aging through several mechanisms:

  • GLO1 expression changes during aging in humans and mice, with the glyoxalase system becoming less efficient with age

  • Reduced GLO1 activity leads to accumulation of dicarbonyl compounds and increased formation of advanced glycation end products (AGEs)

  • In skin, GLO1 protects keratinocyte progenitors during chronological aging and photoaging

  • Animal models demonstrate that GLO1 overexpression extends lifespan, while GLO1 knockdown results in hypersensitivity to MG and shortened lifespan

Researchers investigating this relationship should employ longitudinal study designs to capture the progressive nature of age-related GLO1 decline. For instance, a recent study with Glo1 heterozygous knockdown mice (Glo1+/-) with approximately 50% gene expression demonstrated age-dependent metabolic phenotypes including obesity, hyperglycemia, and dyslipidemia . These phenotypes typically manifested after 14 weeks of age, highlighting the importance of extended observation periods in aging studies .

What are the most effective methods for studying GLO1 knockdown effects in human cell models?

For studying GLO1 knockdown effects in human cell models, researchers should implement a comprehensive experimental approach:

  • siRNA-mediated knockdown: This technique has been successfully employed in human aortic endothelial cells (HAECs) to reduce GLO1 expression under hyperglycemic conditions . siRNA targeting specific GLO1 sequences provides transient but significant reduction in GLO1 activity.

  • Validation of knockdown efficiency:

    • Western blotting to verify protein reduction

    • qPCR to confirm mRNA downregulation

    • Direct enzyme activity assays to confirm functional consequences

    • Measurement of MG concentrations in both cells and culture medium

  • Downstream analyses:

    • Proteomic analysis to identify differentially abundant proteins

    • Detection of MG-glycated protein targets

    • Assessment of functional markers specific to the cell type (e.g., endothelial function markers for HAECs)

    • Evaluation of apoptosis markers

For endothelial cell studies specifically, researchers should examine collagen content in cell culture supernatant, endothelin-1 levels, eNOS phosphorylation, and expression of inflammatory markers (ICAM-1, VCAM-1, MCP-1) .

How can researchers effectively measure methylglyoxal-derived advanced glycation end products (MG-AGEs) in experimental systems?

Accurate measurement of MG-AGEs requires a multifaceted approach combining analytical chemistry and immunological techniques:

  • Liquid chromatography-tandem mass spectrometry (LC-MS/MS):

    • Provides the most specific and sensitive detection of MG-derived adducts

    • Allows quantification of specific AGE species like Nε-carboxymethyl-lysine (CML)

    • Requires hydrolysis of proteins to amino acids followed by derivatization

  • Immunological detection:

    • Western blotting with antibodies specific for MG-derived epitopes

    • Immunohistochemistry for tissue localization of AGEs

    • ELISA for quantification in biological fluids

  • Fluorescence spectroscopy:

    • Measurement of AGE-specific fluorescence (excitation ~370 nm, emission ~440 nm)

    • Simple screening method but less specific than LC-MS/MS

When interpreting results, researchers should recognize that MG-AGEs may not always correlate with phenotypic changes. For example, in Glo1+/- mice, MG-derived AGEs were increased only in male skeletal muscle but not in female tissues, despite females exhibiting more severe metabolic phenotypes . This highlights the importance of considering AGE-independent mechanisms of GLO1 function in experimental design and data analysis.

What animal models provide the most translatable insights for human GLO1 research?

For translatable insights in GLO1 research, researchers should carefully select animal models that reflect human physiology and pathophysiology:

  • Heterozygous Glo1 knockdown (Glo1+/-) mice:

    • More physiologically relevant than complete knockout

    • Exhibit 45-65% reduction in Glo1 enzymatic activity, similar to variations seen in human populations

    • Develop obesity, hyperglycemia, and dyslipidemia in an age and sex-dependent manner

    • Allow longitudinal studies of metabolic phenotypes

  • Experimental considerations for animal studies:

    • Include both sexes in study design, as Glo1+/- females show more severe glucose intolerance and altered lipid metabolism compared to males

    • Conduct longitudinal studies from young to aged animals, as phenotypes typically manifest after 14 weeks of age

    • Analyze multiple tissues (liver, adipose, muscle, kidney, aorta) to capture tissue-specific effects

    • Assess both AGE-dependent and AGE-independent mechanisms

  • Validation in human tissues:

    • Confirm key findings from animal models in human tissue samples when possible

    • Compare expression patterns across species to identify conserved and divergent aspects of GLO1 function

For integrative analyses, researchers should consider transcriptomic profiling across tissues to identify pathway perturbations and potential regulatory genes that explain the connection between Glo1 and human metabolic traits and diseases .

How do sex differences impact GLO1 function and influence experimental outcomes in GLO1 research?

Sex differences significantly impact GLO1 function and experimental outcomes, necessitating careful consideration in research design:

Recent studies with Glo1 heterozygous knockdown (Glo1+/-) mice revealed pronounced sex-dependent phenotypes:

  • Female-specific phenotypes:

    • More severe glucose intolerance at 23 and 33 weeks of age

    • Elevated plasma triglycerides

    • Significant perturbations in genes involved in adipogenesis, PPARγ signaling, insulin signaling, and fatty acid metabolism in liver and adipose tissues

  • Male-specific phenotypes:

    • Increased skeletal muscle mass and visceral adipose depots

    • Changes in lipid metabolism pathways

    • Increased methylglyoxal-derived AGEs specifically in skeletal muscle

  • Transcription factor analysis identified female-biased regulators:

    • Hnf4a (across all tissues)

    • Arntl (in aorta, liver, and kidney)

These findings underscore the necessity of including both sexes in GLO1 research and conducting sex-stratified analyses. Researchers should:

  • Design studies with adequate power to detect sex-specific effects

  • Report sex-specific data even when differences are not observed

  • Investigate potential hormonal influences on GLO1 expression and function

  • Consider sex-specific molecular mechanisms underlying observed phenotypes

The sex-dependent role of GLO1 highlights the importance of personalized approaches in translating GLO1 research into clinical applications .

What are the most significant discrepancies in the literature regarding GLO1 function in human diseases, and how might researchers address these contradictions?

Several significant discrepancies exist in the GLO1 literature that researchers should address:

  • Contradiction regarding GLO1 role in cancer:

    • Some studies identify GLO1 as a potential anticancer target due to its overexpression in multiple cancer types

    • Conversely, other research suggests GLO1 deficiency might promote carcinogenesis through increased MG-induced DNA damage

  • Inconsistencies in AGE-dependent vs. AGE-independent mechanisms:

    • Traditional view emphasizes AGE formation as the primary mechanism of GLO1-related pathology

    • Recent evidence from Glo1+/- mice shows broad metabolic phenotypes without consistent changes in AGE levels across tissues

    • Transcriptional profiles suggest altered glucose and lipid metabolism may be partially explained by alternative detoxification of methylglyoxal to metabolites such as pyruvate

  • Discrepancies in tissue-specific effects:

    • Variable GLO1 expression and function across tissues complicates interpretation of systemic phenotypes

    • Contradictory reports on whether endothelial or parenchymal GLO1 is more critical in vascular pathology

To address these contradictions, researchers should:

  • Employ systems biology approaches integrating transcriptomics, proteomics, and metabolomics

  • Develop tissue-specific conditional GLO1 knockout models

  • Use stable isotope tracing to map metabolic flux through the glyoxalase system

  • Integrate findings with human genome-wide association studies (GWAS) to provide additional mechanistic and clinical insights

  • Report experimental conditions thoroughly to enable accurate cross-study comparisons

How can researchers distinguish between direct effects of GLO1 modulation and secondary consequences of altered methylglyoxal metabolism?

Distinguishing direct GLO1 effects from secondary consequences requires sophisticated experimental approaches:

  • Temporal analysis:

    • Examine early molecular changes following GLO1 modulation before secondary adaptations occur

    • Use inducible systems for controlled timing of GLO1 modulation

    • Monitor time-course of MG accumulation relative to observed phenotypes

  • Pathway dissection strategies:

    • Employ metabolite rescue experiments (e.g., testing if pyruvate supplementation can reverse effects of GLO1 inhibition)

    • Use specific inhibitors targeting distinct steps in MG metabolism

    • Perform parallel experiments modulating alternative MG detoxification pathways

  • Molecular approaches:

    • Identify direct MG modification targets using mass spectrometry-based proteomic approaches

    • Employ CRISPR-based mutagenesis of potential MG target sites in specific proteins

    • Use proximity labeling methods to identify direct GLO1 protein-protein interactions

  • Computational methods:

    • Construct network models integrating transcriptomic, proteomic, and metabolomic data

    • Apply causal inference algorithms to distinguish primary from secondary effects

    • Perform comparative analysis across multiple GLO1 modulation models

Recent research indicates that altered glucose and lipid metabolism in GLO1-deficient models may be partially explained by alternative detoxification of methylglyoxal to metabolites such as pyruvate . This highlights the importance of comprehensive metabolic profiling when studying GLO1 function.

What technical limitations must researchers address when studying GLO1 in human samples?

Researchers face several technical challenges when studying GLO1 in human samples:

  • Sample preservation challenges:

    • MG is highly reactive with half-life of minutes, requiring rapid sample processing

    • GLO1 activity can be affected by sample storage conditions and freeze-thaw cycles

    • AGEs continue forming ex vivo unless properly inhibited

  • Analytical challenges:

    • Difficulty in standardizing GLO1 activity assays across laboratories

    • Cross-reactivity issues with antibodies detecting specific AGE epitopes

    • Low abundance of specific MG-derived adducts requires highly sensitive detection methods

  • Methodological standardization:

    • Variation in protocols for measuring GLO1 activity and MG levels

    • Differences in normalization strategies (per protein, per cell, per tissue weight)

    • Inconsistent definition of "normal" vs. "reduced" GLO1 activity

  • Clinical sample limitations:

    • Access to relevant human tissues (especially for cardiovascular and neurological studies)

    • Ethical constraints on longitudinal sampling

    • Comorbidities and medications affecting GLO1 activity in patient samples

To address these challenges, researchers should:

  • Develop standardized protocols for sample collection and processing

  • Use multiple complementary methods to assess GLO1 activity and MG levels

  • Establish reference ranges for GLO1 activity in different human tissues

  • Consider the impact of demographic factors (age, sex, ethnicity) on GLO1 function

What experimental controls are essential when investigating the effects of GLO1 modulation?

When investigating GLO1 modulation effects, researchers must implement rigorous controls:

  • Essential genetic controls:

    • For knockdown studies: use scrambled siRNA sequences with similar GC content

    • For overexpression studies: empty vector controls expressing the same antibiotic resistance

    • For CRISPR-based approaches: non-targeting guide RNAs and verification of off-target effects

  • Biochemical validation controls:

    • Direct measurement of GLO1 enzyme activity to confirm functional consequences of genetic manipulation

    • Quantification of MG levels to verify the expected biochemical effect

    • Assessment of glutathione levels, as GSH is a required cofactor for GLO1

  • Phenotypic specificity controls:

    • Rescue experiments using MG scavengers (e.g., aminoguanidine)

    • Complementation with exogenous GLO1 to verify phenotype reversibility

    • Parallel knockdown of related enzymes (e.g., GLO2) to distinguish pathway-specific effects

  • Time-course and dosage controls:

    • In GLO1 inhibitor studies, include dose-response analyses

    • For genetic models, use heterozygous (Glo1+/-) in addition to homozygous knockouts

    • Monitor phenotypes longitudinally, as some effects may only manifest at specific ages

  • Multi-tissue analysis:

    • Examine effects across multiple tissues to distinguish systemic from tissue-specific consequences

    • Account for potential compensatory mechanisms in chronic GLO1 deficiency models

How can researchers integrate multi-omics approaches to gain comprehensive insights into GLO1 biology?

Integrating multi-omics approaches offers powerful insights into GLO1 biology:

  • Comprehensive multi-omics strategy:

    • Transcriptomics: Identify altered gene expression patterns and regulatory networks

    • Proteomics: Detect protein abundance changes and post-translational modifications

    • Metabolomics: Map changes in metabolic pathways affected by GLO1 modulation

    • Glycomics: Characterize alterations in protein glycation patterns

  • Integration methods:

    • Pathway enrichment analysis across different omics layers

    • Network-based approaches identifying perturbed molecular interactions

    • Time-resolved multi-omics to capture dynamic responses to GLO1 modulation

    • Machine learning algorithms to identify patterns across complex datasets

  • Translational integration:

    • Correlation of experimental omics data with human GWAS results

    • Integration with clinical parameters from patient cohorts

    • Mapping of tissue-specific transcriptomic signatures to human disease phenotypes

  • Practical implementation:

    • Design experiments with matched samples across all omics platforms

    • Include standardized quality control samples

    • Apply appropriate normalization strategies for cross-platform integration

    • Use established bioinformatic pipelines while remaining aware of their limitations

This integrated approach has revealed that GLO1 reduction perturbs metabolic health through both MG-dependent and independent mechanisms, with pronounced sex differences in underlying molecular pathways . Multi-omics integration provides a systems-level understanding that cannot be achieved through any single approach.

What novel approaches are being developed to target the GLO1 pathway for therapeutic applications?

Several innovative approaches are emerging for therapeutic targeting of the GLO1 pathway:

  • Structure-based drug design:

    • Crystal structures of human GLO1 and its complex with inhibitors like TLSC702 reveal specific binding modes

    • Inhibitors coordinate with Zn²⁺ in the active site through carboxyl oxygen atoms

    • Van der Waals interactions with hydrophobic residues stabilize inhibitor binding

    • This structural information enables rational design of more selective GLO1 inhibitors

  • Tissue-specific GLO1 modulation:

    • Targeted delivery systems to modulate GLO1 in specific tissues

    • Cell type-specific promoters for genetic interventions

    • Nanoparticle-based approaches for organ-specific drug delivery

  • Combined pathway interventions:

    • Dual targeting of GLO1 and alternative MG detoxification pathways

    • Combination of GLO1 modulators with compounds affecting related metabolic pathways

    • Adjunctive therapies addressing downstream consequences of altered GLO1 function

  • Non-pharmacological approaches:

    • Dietary interventions that naturally upregulate GLO1 activity

    • Lifestyle modifications that reduce MG production (e.g., exercise regimens)

    • Nutraceutical compounds that influence the glyoxalase system

Researchers should carefully consider sex differences when developing therapeutic strategies, as recent findings demonstrate significant sex-dependent effects of GLO1 modulation on metabolism . Additionally, age-dependent effects suggest that intervention timing may be critical for therapeutic efficacy.

How is GLO1 research contributing to our understanding of cellular stress response networks?

GLO1 research provides critical insights into cellular stress response networks:

  • Integration with oxidative stress pathways:

    • GLO1 function is closely linked to glutathione (GSH) metabolism

    • Cross-talk between dicarbonyl stress and oxidative stress response elements

    • Synergistic effects of combined oxidative and dicarbonyl stress in cellular damage

  • Connection to proteostasis networks:

    • MG-modified proteins may trigger unfolded protein responses

    • GLO1 dysfunction affects protein quality control mechanisms

    • Potential links between GLO1 and autophagy pathways

  • Metabolic stress integration:

    • GLO1 as a sensor for glycolytic flux and metabolic state

    • Role in adaptive responses to metabolic perturbations

    • Connections between GLO1 and cellular energy sensing networks

  • Transcriptional regulation networks:

    • Identification of transcription factors like Hnf4a and Arntl as regulators influenced by GLO1 status

    • GLO1 modulation affects multiple stress-responsive transcriptional programs

    • Potential epigenetic mechanisms linking GLO1 function to long-term cellular adaptations

Recent transcription factor analyses from tissue-specific gene expression data identified factors involved in cardiometabolic diseases such as Hnf4a (across multiple tissues) and Arntl (in aorta, liver, and kidney) as female-biased regulators whose targets are altered in response to GLO1 reduction . These findings place GLO1 within broader regulatory networks governing cellular stress responses.

What role might GLO1 play in emerging research on cellular senescence and aging biology?

GLO1 plays a multifaceted role in cellular senescence and aging biology research:

  • Senescence mechanisms:

    • MG accumulation due to decreased GLO1 activity may trigger senescence-associated secretory phenotype (SASP)

    • GLO1 dysfunction contributes to telomere attrition and DNA damage responses

    • Potential role in mitochondrial dysfunction linked to cellular senescence

  • Tissue-specific aging phenotypes:

    • In skin, GLO1 protects keratinocyte progenitors during chronological aging and photoaging

    • Accumulation of glycated modified proteins is associated with skin aging process

    • GLO1 expression changes during aging along the lifespan in humans and mice

  • Lifespan regulation:

    • GLO1 overexpression induces prolonged lifespan in model organisms, partially explained by lower content in glycated mitochondrial proteins

    • Knockdown of GLO1 leads to hypersensitivity to MG and shorter lifespan

    • The glyoxalase system appears to become less efficient with age

  • Interventional opportunities:

    • GLO1 as a potential target for "geroprotective" interventions

    • Connections to established longevity pathways (insulin/IGF-1, mTOR)

    • Role in mediating benefits of dietary restriction and other lifespan-extending interventions

Researchers investigating GLO1 in aging should consider both direct effects on cellular function and indirect consequences through systemic metabolism, as GLO1 reduction perturbs metabolic health in age-dependent ways that may contribute to organismal aging .

How do genetic variations in human GLO1 correlate with disease susceptibility and progression?

Genetic variations in human GLO1 show important correlations with disease susceptibility:

  • Polymorphisms affecting GLO1 function:

    • Several single nucleotide polymorphisms (SNPs) impact GLO1 enzymatic activity

    • The C419A polymorphism (Ala111Glu) affects enzyme kinetics and stability

    • Copy number variations influence GLO1 expression levels

  • Disease associations:

    • Diabetes and diabetic complications: GLO1 variants correlate with microvascular and macrovascular complication risk

    • Cardiovascular disease: Polymorphisms associated with coronary artery disease independent of diabetes status

    • Neuropsychiatric conditions: GLO1 was identified as a possible target for anxiety and depression treatment

  • Methodological approaches for genetic studies:

    • Candidate gene association studies examining specific GLO1 variants

    • Genome-wide association studies (GWAS) identifying GLO1 locus associations

    • Integration of tissue-specific transcriptomic signatures with human GWAS to provide mechanistic insights

  • Pharmacogenomic implications:

    • Genetic variations may predict response to GLO1-targeting therapies

    • Potential for personalized medicine approaches based on GLO1 genotype

    • Sex-specific genetic effects requiring stratified analysis approaches

When designing genetic association studies, researchers should consider the age and sex-dependent nature of GLO1-related phenotypes, as demonstrated in experimental models where metabolic changes typically manifest after 14 weeks of age and show pronounced sex differences .

What biomarkers derived from GLO1 research have potential clinical utility?

Several biomarkers derived from GLO1 research show promise for clinical applications:

  • Direct GLO1-related biomarkers:

    • GLO1 enzyme activity in blood cells (erythrocytes, leukocytes)

    • GLO1 protein levels in accessible tissues

    • GLO1 genetic variants as predictive markers

  • Substrate/product biomarkers:

    • Plasma or urinary methylglyoxal concentrations

    • S-D-lactoylglutathione levels

    • D-lactate as an end product of the glyoxalase pathway

  • MG-derived modification biomarkers:

    • MG-hydroimidazolone (MG-H1) in proteins

    • Nε-carboxymethyl-lysine (CML) modified proteins

    • MG-derived DNA adducts

  • Downstream functional biomarkers:

    • Expression of ICAM-1, VCAM-1 and MCP-1 as markers of endothelial inflammation

    • Endothelin-1 levels reflecting endothelial dysfunction

    • Collagen profiles indicative of vascular remodeling

For clinical implementation, researchers should:

  • Establish standardized assay protocols with defined reference ranges

  • Validate biomarkers in large, diverse patient cohorts

  • Assess their predictive value for specific disease outcomes

  • Determine whether sex-specific reference ranges are needed, given the observed sexual dimorphism in GLO1 function

How can findings from basic GLO1 research be effectively translated into clinical applications?

Translating basic GLO1 research into clinical applications requires a strategic approach:

  • Target validation strategies:

    • Confirmation of GLO1 pathway importance in human tissues

    • Correlation of GLO1 activity with disease severity in patient cohorts

    • Integration of experimental findings with human genetic data

  • Therapeutic development considerations:

    • Tissue-specific delivery of GLO1 modulators

    • Addressing potential side effects of systemic GLO1 inhibition

    • Personalized approaches considering sex differences in GLO1 function

  • Clinical trial design elements:

    • Stratification by GLO1 genotype and baseline activity

    • Sex-specific analysis of treatment effects

    • Age-appropriate dosing considerations

    • Biomarker monitoring to confirm target engagement

  • Implementation research:

    • Translation of laboratory GLO1 assays to clinical diagnostics

    • Development of point-of-care testing for MG and related biomarkers

    • Integration of GLO1-based biomarkers into risk assessment algorithms

Recent research highlighting age and sex-dependent effects of GLO1 modulation underscores the importance of personalized approaches . The integration of tissue-specific transcriptomic signatures with human GWAS provides additional mechanistic and clinical insights for translational research . Furthermore, understanding the connection between GLO1 and multiple human metabolic traits and diseases can guide targeted therapeutic development.

What are the most promising unexplored areas for future GLO1 research?

Several promising areas remain unexplored in GLO1 research:

  • Intercellular GLO1 dynamics:

    • Potential for intercellular transport of GLO1 or its products

    • Role of extracellular vesicles in transporting GLO1 or MG-modified proteins

    • Communication between tissues with different GLO1 expression profiles

  • Developmental aspects:

    • GLO1 function during embryonic and postnatal development

    • Epigenetic programming of GLO1 expression

    • Intergenerational effects of parental GLO1 dysfunction

  • Environmental interactions:

    • Impact of environmental toxicants on GLO1 function

    • Dietary components affecting GLO1 activity and expression

    • Microbiome interactions with host GLO1 system

  • Novel regulatory mechanisms:

    • Non-coding RNAs regulating GLO1 expression

    • Post-translational modifications affecting GLO1 activity

    • Subcellular compartmentalization of GLO1 function

  • Integration with emerging biological paradigms:

    • Role in cellular phase separation and biomolecular condensates

    • Connections to circadian rhythms, particularly through Arntl regulation

    • Involvement in immunometabolism and trained immunity

Research exploring these areas should incorporate the lessons from recent studies, particularly regarding sex differences and age-dependent effects of GLO1 modulation . Integrative approaches combining multiple omics technologies will be essential for comprehensive understanding of these complex aspects of GLO1 biology.

How might emerging technologies advance our understanding of GLO1 biology?

Emerging technologies offer transformative potential for GLO1 research:

  • Single-cell technologies:

    • Single-cell transcriptomics to reveal cell type-specific GLO1 expression patterns

    • Single-cell proteomics to detect heterogeneous responses to GLO1 modulation

    • Spatial transcriptomics to map GLO1 expression in tissue microenvironments

  • Advanced imaging approaches:

    • Live-cell imaging of GLO1 activity using fluorescent reporters

    • Super-resolution microscopy of GLO1 subcellular localization

    • In vivo imaging of MG dynamics in model organisms

  • Precision genome editing:

    • CRISPR-based screens to identify GLO1 regulators and interactors

    • Base editing to introduce specific GLO1 variants

    • Epigenome editing to modulate GLO1 expression without altering sequence

  • Computational advancements:

    • AI-driven prediction of MG modification sites

    • Network modeling of GLO1 in cellular stress responses

    • Multi-scale simulation of GLO1 effects from molecular to organismal levels

  • Microfluidic systems:

    • Organ-on-chip models incorporating GLO1 pathway components

    • High-throughput screening for GLO1 modulators

    • Miniaturized assays for GLO1 activity in limited samples

Researchers implementing these technologies should consider the differential effects of GLO1 modulation across tissues, sexes, and ages , designing experiments that capture this biological complexity while leveraging technological advantages for deeper mechanistic insights.

What interdisciplinary collaborations would most benefit the advancement of GLO1 research?

Advancing GLO1 research would benefit significantly from strategic interdisciplinary collaborations:

  • Basic science collaborations:

    • Biochemists and structural biologists to elucidate GLO1 molecular mechanisms

    • Glycobiologists to investigate MG-derived modifications

    • Systems biologists for network-level analysis of GLO1 pathways

  • Clinical research partnerships:

    • Endocrinologists studying metabolic disorders

    • Cardiologists investigating vascular complications

    • Dermatologists researching skin aging processes

    • Oncologists exploring GLO1 in cancer biology

  • Technological collaborations:

    • Analytical chemists for advanced MG detection methods

    • Bioengineers developing targeted delivery systems

    • Computer scientists for AI-based data integration

    • Bioinformaticians for multi-omics data analysis

  • Translational research teams:

    • Drug development specialists for GLO1-targeting therapeutics

    • Biomarker development experts

    • Regulatory affairs consultants for clinical translation

    • Health economists for cost-effectiveness analysis

  • Public health perspectives:

    • Epidemiologists studying population-level GLO1 variation

    • Nutrition scientists investigating dietary impacts on GLO1

    • Environmental health researchers examining toxicant effects

Successful collaborations should acknowledge the complex, context-dependent nature of GLO1 biology, particularly the pronounced sex differences and age-dependent effects observed in recent research . Interdisciplinary teams can address these complexities more effectively than siloed research approaches.

What high-throughput screening approaches can identify novel modulators of GLO1 activity?

Effective high-throughput screening for GLO1 modulators requires sophisticated methodological approaches:

  • Biochemical assay platforms:

    • Fluorescence-based GLO1 activity assays monitoring S-D-lactoylglutathione formation

    • Coupling GLO1 activity to secondary enzymes for amplified detection

    • Label-free technologies measuring direct enzyme-substrate interactions

  • Cell-based screening systems:

    • Reporter cell lines with fluorescent/luminescent readouts linked to GLO1 activity

    • High-content imaging of MG-derived adduct formation

    • Phenotypic screens measuring biological consequences of GLO1 modulation

  • In silico screening approaches:

    • Structure-based virtual screening using GLO1 crystal structures

    • Molecular docking targeting the Zn²⁺ coordination site

    • Machine learning models predicting GLO1-modulating properties

  • Screening library considerations:

    • Natural product libraries for bioactive GLO1 modulators

    • Fragment-based approaches for novel scaffold identification

    • Repurposing libraries of clinically approved compounds

  • Validation cascade:

    • Secondary biochemical assays with purified human GLO1

    • Cellular target engagement confirmation

    • Specificity profiling against related enzymes

    • Sex-specific cellular models to capture dimorphic responses

Researchers should incorporate learnings from crystal structures of human GLO1 in complex with inhibitors like TLSC702, which reveal specific binding modes coordinating with Zn²⁺ through carboxyl oxygen atoms while establishing van der Waals interactions with hydrophobic residues .

How can computational modeling advance the design of selective GLO1 modulators?

Computational modeling offers powerful approaches for designing selective GLO1 modulators:

  • Structure-based design strategies:

    • Molecular dynamics simulations of GLO1-inhibitor interactions

    • Quantum mechanical calculations of zinc coordination chemistry

    • Analysis of protein flexibility and cryptic binding sites

  • Ligand-based approaches:

    • Pharmacophore modeling based on known GLO1 inhibitors like TLSC702

    • Quantitative structure-activity relationship (QSAR) models

    • Machine learning prediction of binding affinities

  • Advanced computational methods:

    • Free energy perturbation calculations for binding affinity prediction

    • Metadynamics for exploring conformational landscapes

    • Fragment-based computational screening

  • System-level modeling:

    • Pathway simulations predicting consequences of GLO1 modulation

    • Physiologically-based pharmacokinetic (PBPK) modeling

    • Sex-specific models accounting for dimorphic responses

  • Practical implementation:

    • Integration of crystallographic data on human GLO1-inhibitor complexes

    • Accounting for the role of the Zn²⁺ coordination in the active site

    • Modeling hydrophobic interactions with specific residues

Crystal structures of human GLO1 reveal that inhibitors coordinate with Zn²⁺ through carboxyl oxygen atoms while establishing van der Waals interactions with hydrophobic residues . This structural information forms the foundation for rational structure-based drug design efforts. Additionally, computational models should account for the observed sex differences in GLO1 function to predict potential sex-specific responses to GLO1 modulators .

What novel analytical techniques are advancing the detection and quantification of GLO1 substrates and products?

Novel analytical techniques are revolutionizing the detection and quantification of GLO1 substrates and products:

  • Advanced mass spectrometry approaches:

    • Stable isotope dilution LC-MS/MS for absolute quantification of MG

    • Ion mobility-mass spectrometry for improved separation of isomeric species

    • MALDI imaging mass spectrometry for spatial distribution of MG adducts

    • Targeted proteomics for specific MG-modified peptides

  • Real-time monitoring technologies:

    • Fluorescent probes specifically reacting with MG

    • Genetically encoded biosensors for intracellular MG detection

    • Electrochemical sensors for continuous monitoring

    • Microfluidic devices for dynamic measurement of glyoxalase activity

  • Multi-analyte detection systems:

    • Multiplex assays simultaneously quantifying MG, glyoxal, and 3-deoxyglucosone

    • Combined detection of free MG and protein-bound MG-derived adducts

    • Integrated analysis of glyoxalase system components (substrates, intermediates, products)

  • Enhanced sensitivity methods:

    • Chemical derivatization strategies improving MG detection limits

    • Nanoparticle-based signal amplification

    • Digital droplet approaches for single-molecule sensitivity

  • Sample preparation innovations:

    • Rapid quenching techniques preventing ex vivo MG formation

    • Selective extraction methods for enriching MG and MG-modified molecules

    • Automated sample preparation minimizing human error

Product Science Overview

Structure and Function

Glyoxalase-I is a 21 kDa isomerase enzyme that catalyzes the formation of S-D-lactoylglutathione from the hemimercaptal adduct formed spontaneously between methylglyoxal and reduced glutathione (GSH) . This reaction is the first step in the glyoxalase pathway, which ultimately converts methylglyoxal into D-lactate, a less harmful compound .

Importance in Cellular Metabolism

The glyoxalase pathway, which includes Glyoxalase-I and Glyoxalase-II, is evolutionarily conserved and plays a significant role in maintaining cellular homeostasis by detoxifying methylglyoxal . By converting methylglyoxal to D-lactate, Glyoxalase-I helps protect cells from glycation and oxidative stress, which can damage proteins, nucleic acids, and lipids .

Recombinant Human Glyoxalase-I

Recombinant human Glyoxalase-I is produced using E. coli expression systems. The recombinant protein typically includes an N-terminal Met and a 6-His tag for purification purposes . It is often used in research to study the enzyme’s function, structure, and role in various diseases, including diabetes and cancer .

Applications in Research

Recombinant Glyoxalase-I is valuable in biochemical research for several reasons:

  • Studying Enzyme Kinetics: Researchers use it to understand the enzyme’s catalytic mechanisms and substrate specificity.
  • Drug Development: It serves as a target for developing inhibitors that could potentially treat diseases associated with high levels of methylglyoxal.
  • Disease Research: Understanding how Glyoxalase-I functions can provide insights into its role in conditions like diabetes, where methylglyoxal levels are elevated .

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 2025 TheBiotek. All Rights Reserved.