GALK1 Human

Galactokinase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Enzyme Structure

GALK1 belongs to the GHMP (galactokinase, homoserine kinase, mevalonate kinase, phosphomevalonate kinase) superfamily. Its structure includes:

  • N-terminal domain: Mixed β-sheets and α-helices.

  • C-terminal domain: Anti-parallel β-sheets and α-helices.

  • Active site: Binds ATP and galactose, with conserved residues:

    • Asp-186: Abstracts a proton from galactose, enabling nucleophilic attack on ATP’s γ-phosphorus.

    • Arg-37: Stabilizes Asp-186 in its anionic form, essential for catalysis .

DomainKey FeaturesRole
N-terminalMixed β-sheets, α-helicesATP binding interface
C-terminalAnti-parallel β-sheets, α-helicesGalactose substrate recognition

Metabolic Role and Pathway

GALK1 initiates the Leloir pathway, converting dietary galactose (e.g., from lactose) into glucose. Key steps include:

  1. GALK1: Galactose → Gal-1-P (using ATP).

  2. Galactose-1-phosphate uridylyltransferase (GALT): Gal-1-P → UDP-galactose.

  3. UDP-galactose 4-epimerase: UDP-galactose ↔ UDP-glucose .

StepEnzymeProduct
1GALK1Galactose-1-phosphate
2Galactose-1-phosphate uridylyltransferaseUDP-galactose
3UDP-galactose 4-epimeraseUDP-glucose

Genetic Variants and Clinical Implications

GALK1 deficiency (galactosemia type II) arises from mutations impairing enzyme activity. Over 30 pathogenic variants have been identified, including:

  • Pro28Thr (c.82C>A): Founder mutation in Roma populations.

  • Ala198Val (c.593C>T): Common in Japanese/Korean populations.

  • Q382X: Nonsense mutation truncating the enzyme .

MutationPopulationPhenotype
Pro28ThrRomaCongenital cataracts
Ala198ValJapanese/KoreanPresenile cataracts, hypergalactosemia
Q382XCosta RicanSevere enzyme deficiency

Clinical Findings:

  • Cataracts: Primary manifestation due to galactitol accumulation in lens cells, causing osmotic damage .

  • Developmental Delays: Reported in some cases, though causality remains debated .

  • Biochemical Markers:

    • Galactose: Elevated in blood/plasma.

    • Galactitol: Urinary levels >2500 mmol/mol creatinine pre-treatment .

Enzyme Activity in Health and Disease

ParameterHealthy ControlsGALK1-Deficient Patients
GALK1 Activity1.0–2.7 μmol/g Hgb/hr<20% of normal
Galactose LevelsNormalHypergalactosemia
Galactitol LevelsUndetectableElevated (urinary)

Inhibitor Development

Structure-based optimization of dihydropyrimidine inhibitors (e.g., GALK1 inhibitors) aims to reduce Gal-1-P accumulation in classic galactosemia. Key findings:

  • Biochemical Efficacy: IC₅₀ < 100 nM in vitro.

  • Cellular Activity: Prevent Gal-1-P buildup in patient-derived cells at low micromolar concentrations.

  • Pharmacokinetics: Suitable for preclinical testing in rodent models .

Diagnostic and Genetic Testing

  • Newborn Screening (NBS): Identifies GALK1 deficiency via enzyme activity assays or genetic testing.

  • Genetic Testing:

    • Mutation Detection: Sequencing for founder mutations (e.g., Pro28Thr, Ala198Val).

    • Enzyme Assays: LC-MS/MS for GALK1 activity in erythrocytes .

Product Specs

Introduction
The GALK1 enzyme plays a crucial role in the initial step of the galactose metabolism pathway, converting ATP and D-galactose into ADP and alpha-D-galactose 1-phosphate. A deficiency in GALK1 leads to galactosemia II, an autosomal recessive disorder characterized by congenital cataracts in infancy and presenile cataracts in adults. The cataract formation is attributed to the accumulation of galactitol in the lenses.
Description
Recombinant Human GALK1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 412 amino acids (specifically, amino acids 1 to 392). It possesses a molecular weight of 44.4 kDa. The GALK1 protein has a 20 amino acid His-Tag fused to its N-terminus and undergoes purification using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, colorless solution that has been filtered for sterility.
Formulation
This solution contains 0.5mg/ml of Human GALK1 in a buffer consisting of 20% glycerol, 0.1M NaCl, and 1mM DTT.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For extended storage, it is recommended to freeze the product at -20°C. To further enhance long-term stability, consider adding a carrier protein (0.1% HSA or BSA). Repeated freezing and thawing should be avoided.
Purity
The purity of the GALK1 protein is determined to be greater than 95.0% based on SDS-PAGE analysis.
Synonyms
Galactose kinase, GK1, GALK, EC 2.7.1.6, GALK1.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MAALRQPQVA ELLAEARRAF REEFGAEPEL AVSAPGRVNL IGEHTDYNQG LVLPMALELM TVLVGSPRKD GLVSLLTTSE GADEPQRLQF PLPTAQRSLE PGTPRWANYV KGVIQYYPAA PLPGFSAVVV SSVPLGGGLS SSASLEVATY TFLQQLCPDS GTIAARAQVC QQAEHSFAGM PCGIMDQFIS LMGQKGHALL IDCRSLETSL VPLSDPKLAV LITNSNVRHS LASSEYPVRR RQCEEVARAL GKESLREVQL EELEAARDLV SKEGFRRARH VVGEIRRTAQ AAAALRRGDY RAFGRLMVES HRSLRDDYEV SCPELDQLVE AALAVPGVYG SRMTGGGFGG CTVTLLEASA APHAMRHIQE HYGGTATFYL SQAADGAKVL CL.

Q&A

What is the molecular structure of human GALK1 and how does it influence enzyme function?

Human galactokinase 1 (GALK1) belongs to the GHMP superfamily of kinases which includes galactokinase, homoserine kinase, mevalonate kinase, and phosphomevalonate kinase . The enzyme follows an ordered ternary complex mechanism with ATP binding occurring first before galactose can bind .

Structurally, certain regions are critical for enzyme function:

  • The active site includes Asp46, which forms direct bonds with galactose through C3-OH and C4-OH interactions

  • Thr352 forms part of a β-sheet structure that controls specificity and activity

  • The enzyme exhibits remarkable intolerance to many single amino acid substitutions, explaining why even minor mutations can cause complete loss of function

Research methodologies to study GALK1 structure include X-ray crystallography, which has revealed binding sites for both substrates and potential inhibitors .

How does GALK1 contribute to normal galactose metabolism in humans?

GALK1 catalyzes the first step in the Leloir pathway of galactose metabolism, converting α-D-galactose to galactose-1-phosphate (Gal-1-P) using ATP as a phosphate donor . This reaction is essential because:

  • It enables further processing of dietary galactose primarily obtained from lactose in dairy products

  • It supports processing of endogenously produced galactose from glycoconjugate turnover

  • It contributes to the production of UDP-galactose needed for galactosylation of proteins and lipids

In erythrocytes of healthy individuals, GALK1 activity measured by LC-MS/MS assays ranges from 1.0–2.7 μmol·(g Hgb)−1·hr−1, with a mean activity of 1.8 ± 0.43 μmol·(g Hgb)−1·hr−1 . This enzymatic activity is critical for preventing accumulation of galactose and its conversion to alternative metabolites like galactitol.

What methodologies are used to assess GALK1 enzyme activity in clinical and research settings?

Several complementary approaches are employed to evaluate GALK1 functionality:

  • Biochemical enzyme activity assays:

    • Liquid chromatography-tandem mass spectrometry (LC-MS/MS) to quantify enzyme activity in erythrocytes, with normal range of 1.0–2.7 μmol·(g Hgb)−1·hr−1

    • Radioisotope-based assays tracking the conversion of labeled galactose to galactose-1-phosphate

  • Genetic analysis approaches:

    • Sequencing of the GALK1 gene located on chromosome 17q24

    • Identification of pathogenic variants through next-generation sequencing panels

    • In silico prediction tools like PredictSNP to estimate functional impacts of novel variants

  • Metabolite measurement:

    • Quantification of galactose, galactitol, and galactonic acid in blood and urine as biomarkers of GALK1 function

    • Monitoring of galactitol levels, which can rise to 2500 mmol/mol creatinine in GALK1-deficient patients prior to dietary intervention

What is the spectrum of pathogenic GALK1 variants and their differential effects on enzyme structure and function?

The mutational spectrum of GALK1 comprises more than 30 disease-causing variants with varying molecular consequences . Research indicates several types of functional impacts:

  • Founder variants with population specificity:

    • NM_000154.1:c.82C>A (p.Pro28Thr): Common in the Roma population

    • NM_000154.1:c.593C>T (p.Ala198Val): The "Osaka variant" prevalent in Japanese and Korean populations

  • Active site disruption:

    • Variants affecting Asp46 (e.g., p.Asp46Asn) disrupt galactose binding, rendering the enzyme catalytically inactive while maintaining protein solubility

    • Experimental studies show that changing Asp46 to alanine produces a soluble but non-functional enzyme

  • Structural integrity variants:

    • Mutations in β-sheet structures controlling specificity and activity, such as those affecting Thr352

    • Variants causing protein instability or improper folding

Methodologically, researchers employ in silico prediction tools like PredictSNP to estimate stability changes, combined with crystallography studies to analyze structural perturbations . Understanding these differential effects guides genetic counseling and informs potential therapeutic approaches.

How do researchers distinguish between pathogenic and benign variants in GALK1 for accurate molecular diagnosis?

Distinguishing pathogenic from benign variants requires a multi-faceted approach:

  • Functional characterization:

    • Enzyme activity assays in patient erythrocytes, with activities below 20% of reference values indicating pathogenicity

    • Expression studies with recombinant variants to assess catalytic activity

  • Computational prediction:

    • Structure-based analysis of amino acid substitutions and their proximity to functional domains

    • Tools like MutationTaster and PredictSNP to predict functional impacts

  • Population frequency analysis:

    • Prevalence in population databases versus disease cohorts

    • Identification of founder mutations with established pathogenicity (e.g., p.Pro28Thr in Roma population)

  • Clinical correlation:

    • Association with typical features like cataracts

    • Biochemical phenotype showing elevated galactitol and galactonic acid

For novel variants, researchers typically require both genetic evidence and functional demonstration of reduced enzyme activity below the 20% threshold to confidently assign pathogenicity .

What are the molecular mechanisms by which GALK1 deficiency leads to cataract formation?

Cataract formation in GALK1 deficiency follows a clear biochemical pathway:

  • Metabolite accumulation:

    • Deficient galactokinase activity prevents conversion of galactose to galactose-1-phosphate

    • This leads to shunting of galactose through alternative pathways

  • Alternative metabolic pathways:

    • Increased flux through the reductive pathway produces elevated galactitol via aldose reductase

    • Enhanced oxidative pathway generates galactonic acid

  • Lens-specific pathology:

    • Galactitol, as a sugar alcohol, accumulates specifically in lens fiber cells

    • Being osmotically active, galactitol draws water into lens cells causing swelling

    • This osmotic stress disrupts lens protein organization and transparency

    • Progressive damage leads to lens opacity (cataract)

Understanding this mechanism has important implications for intervention strategies, including the critical importance of early dietary galactose restriction to prevent irreversible lens damage .

What is the complete phenotypic spectrum of GALK1 deficiency based on registry data?

The GalNet registry data from 53 previously unreported patients across 17 centers in 11 countries provides the most comprehensive phenotypic characterization to date :

Primary manifestations:

  • Cataracts: The predominant and consistent clinical finding

  • Gender distribution: 64.2% male and 35.8% female patients

  • Age range: 1-35 years, with median age of 10.4 years

Additional findings with uncertain attribution:

  • Language delay: Present in 3 of 34 assessed patients (8.8%)

  • Speech disorders: Reported in 3 of 40 patients (7.5%)

  • Neurological symptoms: Isolated reports of movement disorders including tremor, dystonia, and ataxia

  • Developmental: Rare instances of microcephaly

  • Psychiatric manifestations: Attention deficit-hyperactivity disorder and anxiety disorder in some patients

Female gonadal function:

  • Normal puberty in 7 of 8 female patients with reported gonadal follow-up

  • Delayed puberty in one patient

  • No primary ovarian insufficiency reported, contrasting with classic galactosemia

How can GALK1 inhibition serve as a therapeutic strategy for classic galactosemia?

GALK1 inhibition represents a promising "substrate reduction" therapeutic approach for classic galactosemia:

Scientific rationale:

  • Classic galactosemia results from deficiency of galactose-1-phosphate uridylyltransferase (GALT), causing toxic accumulation of galactose-1-phosphate (Gal-1-P)

  • GALK1 inhibition would prevent formation of Gal-1-P, potentially ameliorating disease progression

Supporting experimental evidence:

  • galk1 knockout in Drosophila rescued the galactosemic neurological phenotype

  • galk1 knockdown in GALT-deficient yeast abolished sensitivity to galactose levels

  • Patients with GALK1 deficiency have milder phenotypes than those with classic galactosemia and do not accumulate Gal-1-P

Current research approaches:

  • Fragment-based drug discovery:

    • Crystallography-based screening of fragment libraries

    • Identification of both active site and novel allosteric binding sites

  • Inhibitor categories:

    • ATP-competitive inhibitors (spiro-benzoxazole series)

    • Novel allosteric inhibitors with improved selectivity

    • Merged fragment compounds showing micromolar inhibition without competing with substrates

Research has successfully identified compounds that demonstrate micromolar inhibition of human GALK1 without competing with either substrate (ATP or galactose) and showing good selectivity over homologues like galactokinase 2 and mevalonate kinase . This therapeutic strategy represents a paradigm shift from symptom management to addressing the underlying metabolic dysregulation.

What biochemical markers best reflect GALK1 activity for monitoring disease progression and treatment efficacy?

Multiple biochemical markers provide valuable insights for clinical monitoring of GALK1 deficiency:

  • Direct enzyme activity measurement:

    • Erythrocyte GALK1 activity via LC-MS/MS assays (reference range: 1.0–2.7 μmol·(g Hgb)−1·hr−1)

    • Activities below 20% of reference values considered diagnostic

  • Metabolite quantification:

    • Galactitol levels: The most sensitive biomarker

      • Can reach 2500 mmol/mol creatinine before dietary intervention

      • Significantly decreases following galactose restriction

      • Particularly useful for treatment monitoring

    • Galactose levels: Elevated in untreated patients

      • Reflects dietary exposure and endogenous production

    • Galactonic acid: Elevated from increased oxidative pathway flux

  • Treatment response indicators:

    • Rate and extent of galactitol reduction following dietary galactose restriction

    • Normalization of galactose levels

    • Prevention or stabilization of cataracts

For longitudinal monitoring, urinary galactitol provides the most reliable biomarker for assessing treatment efficacy, with significant decreases observed after introduction of dietary restrictions . Combined assessment of these markers enables comprehensive evaluation of disease status and intervention success.

What are the optimal experimental models for studying GALK1 function and testing potential therapeutic approaches?

Researchers employ diverse experimental models to investigate GALK1, each with specific advantages:

  • Cellular models:

    • Patient-derived fibroblasts: Enable study of endogenous GALK1 function in human cells

    • GALT-deficient cell lines with GALK1 manipulation: Allow investigation of substrate reduction approaches

    • Recombinant expression systems: Useful for characterizing variant effects on enzyme activity

  • Animal models:

    • Drosophila: galk1 knockout flies demonstrated rescue of galactosemic neurological phenotype when combined with GALT deficiency

    • Yeast models: galk1 knockdown in GALT-deficient yeast abolished galactose sensitivity

    • Mammalian models: Provide insights into systemic effects and tissue-specific manifestations

  • Biochemical and structural approaches:

    • Crystallography: Essential for understanding protein structure and inhibitor binding

    • Fragment screening: Identifies starting points for rational drug design

    • Enzyme kinetics assays: Characterize functional impacts of mutations and inhibitors

  • Clinical research platforms:

    • Patient registries: The GalNet registry provides real-world data on disease manifestations and progression

    • Biobanks: Enable access to patient samples for advanced molecular analyses

For therapeutic development, the combination of structural biology approaches with cellular and animal models offers the most comprehensive evaluation pipeline, allowing assessment of both on-target activity and potential off-target effects .

How can researchers effectively design inhibitor studies to target GALK1 with improved selectivity and reduced off-target effects?

Designing selective GALK1 inhibitors requires sophisticated methodological approaches:

  • Structure-based design strategies:

    • Crystallography-based fragment screening: Soaking hundreds of crystals with a custom fragment library identified binding sites beyond the active site

    • Identification of allosteric sites: Eight fragments bound to a hotspot distal from the active site, providing opportunities for highly selective inhibition

    • Fragment merging approach: Combining overlapping fragments improved potency while maintaining selectivity

  • Selectivity assessment:

    • Homologue panel testing: Screening against related enzymes including galactokinase 2 and mevalonate kinase

    • Kinetic mechanism determination: Establishing whether inhibitors are competitive with ATP, galactose, both, or neither

    • Cellular pathway specificity: Evaluating effects on related metabolic pathways

  • Optimization strategies:

    • Structure-activity relationship studies: Systematic modification of lead compounds based on binding interactions

    • Medicinal chemistry refinement: Improving pharmacokinetic properties while maintaining target engagement

    • Computational prediction: Using in silico approaches to predict selectivity profiles before synthesis

Recent achievements include developing micromolar inhibitors of human GALK1 that are not competitive with either substrate (ATP or galactose) and demonstrate good selectivity over related enzymes . This non-competitive mechanism offers advantages over ATP-competitive inhibitors, which may have limited clinical utility due to ATP's ubiquitous role in cellular processes.

What methodology should be employed to accurately diagnose GALK1 deficiency and differentiate it from other forms of galactosemia?

A comprehensive diagnostic approach integrates multiple methodologies:

  • Initial screening and biochemical assessment:

    • Newborn screening: Many patients (35 of 53 in the GalNet registry) were diagnosed through newborn screening programs

    • Galactose levels: Elevated in multiple types of galactosemia

    • Galactitol and galactonic acid: Characteristically elevated in GALK1 deficiency

    • Gal-1-P levels: Typically not accumulated in GALK1 deficiency, unlike classic galactosemia

  • Enzymatic confirmation:

    • GALK1 activity assay: Activity below 20% of reference value indicates deficiency

    • GALT activity testing: Normal in GALK1 deficiency but deficient in classic galactosemia

    • GALE (UDP-galactose-4-epimerase) activity: To rule out type III galactosemia

  • Genetic analysis:

    • Targeted GALK1 sequencing: To identify known pathogenic variants

    • Next-generation sequencing panels: Including all galactosemia-related genes

    • Variant interpretation: Using in silico prediction tools and functional studies for novel variants

  • Clinical correlation:

    • Ophthalmological examination: Assess for cataracts, the primary manifestation

    • Developmental assessment: Evaluate for any associated developmental issues

    • Family history: Identify potential autosomal recessive inheritance pattern

The definitive diagnosis requires demonstration of deficient GALK1 enzyme activity and/or identification of biallelic pathogenic variants in the GALK1 gene . This multi-faceted approach enables accurate differentiation from classic galactosemia (GALT deficiency) and type III galactosemia (GALE deficiency).

What are the current controversies regarding the complete phenotypic spectrum of GALK1 deficiency?

The phenotypic spectrum of GALK1 deficiency remains a subject of scientific debate:

Resolving these controversies requires larger, longitudinal studies with standardized assessment protocols to determine whether reported extra-ophthalmological manifestations are statistically associated with GALK1 deficiency or represent background population frequencies .

How can researchers advance the development of allosteric GALK1 inhibitors from fragment hits to clinical candidates?

The progression from fragment hits to clinical candidates requires a strategic research pipeline:

  • Fragment optimization pathway:

    • Hit validation: Confirming binding through orthogonal techniques beyond crystallography

    • Fragment growing/linking: Systematic expansion of fragments to explore surrounding chemical space

    • Structure-activity relationship studies: Determining which chemical features are critical for activity

  • Mechanism characterization:

    • Allosteric modulation assessment: Understanding how binding to the distal hotspot affects enzyme function

    • Kinetic analysis: Determining inhibition constants and modality (competitive vs. non-competitive)

    • Conformational impacts: Examining how inhibitors affect protein dynamics and substrate binding

  • Pre-clinical development challenges:

    • Pharmacokinetic optimization: Improving absorption, distribution, metabolism, and excretion profiles

    • Target engagement biomarkers: Developing methods to confirm on-target activity in vivo

    • Safety assessment: Evaluating potential off-target effects and toxicity profiles

    • Efficacy models: Establishing appropriate disease models for proof-of-concept studies

  • Translational considerations:

    • Patient stratification strategies: Identifying which classic galactosemia patients might benefit most

    • Biomarker development: Establishing measurable indicators of therapeutic efficacy

    • Combination approaches: Assessing potential synergies with other therapeutic modalities

Current progress includes the successful development of micromolar inhibitors that are not competitive with either substrate and demonstrate good selectivity . Further optimization of these compounds through medicinal chemistry efforts represents the next critical step toward clinical translation.

What research approaches could address the long-term outcomes of early-diagnosed and treated GALK1 deficiency patients?

Systematic investigation of long-term outcomes requires comprehensive research design:

  • Registry expansion and standardization:

    • Extended follow-up protocols: The current GalNet registry includes patients with median age 10.4 years (range 1-35)

    • Standardized assessment tools: Implementing consistent developmental, neurological, and ophthalmological evaluations

    • Quality of life metrics: Incorporating patient-reported outcomes

  • Comparative effectiveness research:

    • Treatment timing analysis: Comparing outcomes between patients diagnosed through newborn screening versus later diagnosis

    • Dietary adherence studies: Assessing the impact of varying levels of galactose restriction

    • Different management approaches: Evaluating various monitoring and intervention protocols

  • Multimodal outcome assessment:

    • Ophthalmological: Long-term visual outcomes and cataract progression/recurrence

    • Developmental: Standardized neurocognitive assessments at key developmental stages

    • Biochemical: Correlation between biomarker control and clinical outcomes

    • Reproductive (for females): Comprehensive assessment of gonadal function and fertility

  • Translational biomarker research:

    • Identification of predictive markers: Determining which early biomarkers correlate with long-term outcomes

    • Development of surrogate endpoints: Establishing validated markers that predict clinical benefit

This comprehensive research approach would address critical knowledge gaps regarding the natural history of treated GALK1 deficiency and provide evidence-based guidance for optimizing management strategies across the lifespan.

Product Science Overview

Structure and Function

Galactokinase 1 is composed of two domains separated by a large cleft. The N-terminal domain consists of five strands of mixed beta-sheet and five alpha-helices, while the C-terminal domain is characterized by two layers of anti-parallel beta-sheets and six alpha-helices . The enzyme binds ATP in a hydrophobic pocket located at the interface of these two domains. This binding is essential for the enzyme’s function, as it facilitates the transfer of a phosphate group from ATP to galactose .

Biological Significance

The primary function of Galactokinase 1 is to convert α-D-galactose to galactose-1-phosphate. This reaction is vital for the proper metabolism of galactose, allowing it to be further processed into glucose-1-phosphate, which can then enter glycolysis or be used in other metabolic pathways . Deficiency in Galactokinase 1 activity can lead to a condition known as galactokinase deficiency or galactosemia type II, characterized by elevated levels of galactose in the blood and urine .

Disease Relevance

Galactokinase deficiency is a rare hereditary disorder with a worldwide incidence of approximately 1 in 1,000,000 . The condition is primarily characterized by the development of cataracts in infancy or childhood due to the accumulation of galactitol, a byproduct of galactose metabolism . Other potential complications include neonatal hypoglycemia, bleeding diathesis, and encephalopathy . Early diagnosis through newborn screening and dietary management can significantly improve outcomes for affected individuals .

Recombinant Galactokinase 1

Recombinant Galactokinase 1 is produced using genetic engineering techniques, where the human GALK1 gene is inserted into a suitable expression system, such as bacteria or yeast. This allows for the production of large quantities of the enzyme for research and therapeutic purposes. Recombinant Galactokinase 1 is used in various studies to understand the enzyme’s structure, function, and role in disease, as well as to develop potential treatments for galactokinase deficiency .

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.