GNMT Human

Glycine N-methyltransferase Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biochemical Functions

GNMT catalyzes the transfer of a methyl group from S-adenosylmethionine (SAM) to glycine, producing sarcosine and S-adenosylhomocysteine (SAH) . This reaction serves two primary roles:

  • Regulation of SAM/SAH Ratio: Maintains methylation capacity by balancing SAM (methyl donor) and SAH (methylation inhibitor) .

  • Detoxification: Binds polycyclic aromatic hydrocarbons (PAHs) and aflatoxins, reducing their carcinogenicity .

Key Kinetic Properties

Speciesk<sub>cat</sub> (min<sup>-1</sup>)K<sub>m</sub> (Glycine)Inhibition by Folate
Human1,2000.8 mMStrong (IC<sub>50</sub>: 2 µM)
Rat8001.2 mMModerate
Mouse9501.0 mMModerate

GNMT also binds 5-methyltetrahydrofolate (5-CH<sub>3</sub>-H<sub>4</sub>PteGlu<sub>5</sub>), linking folate metabolism to methylation .

Cancer Biology

GNMT acts as a tumor suppressor, particularly in hepatocellular carcinoma (HCC):

  • Downregulation in Tumors: GNMT expression is lost in 60-80% of HCC and prostate cancers .

  • Pro-Apoptotic Effects: Transient GNMT expression induces caspase-dependent apoptosis in cancer cells (e.g., HepG2, A549) .

  • Epigenetic Regulation: GNMT deficiency correlates with global DNA hypermethylation and silencing of tumor suppressors .

Genetic Disorders

ConditionMechanismClinical Features
GNMT DeficiencyLoss-of-function mutations (e.g., R239X)Hypermethioninemia, liver fibrosis
Cancer SusceptibilityPolymorphisms (e.g., STRP1) impair detoxificationElevated risk of HCC and prostate cancer

Key Studies

Study FocusFindingsSource
GNMT Knockout MiceDevelop spontaneous HCC; 35-fold elevated SAM levels
Nuclear TranslocationGNMT relocates to nuclei in cancer cells, triggering apoptosis
Detoxification RoleReduces aflatoxin B1-DNA adducts by 70% in vitro
Folate Interaction5-CH<sub>3</sub>-H<sub>4</sub>PteGlu<sub>5</sub> inhibits GNMT, stabilizing cellular folate

Biotechnological Applications

Recombinant human GNMT is produced in E. coli for research and therapeutic exploration:

ApplicationDetailsSource
Enzyme ReplacementPotential therapy for GNMT deficiency (e.g., hypermethioninemia)
Cancer TherapeuticsGNMT delivery via vectors reduces tumor growth in murine models
Biochemical AssaysUsed to study SAM/SAH dynamics and folate metabolism

Product Specs

Introduction
Glycine N-methyltransferase (GNMT) is an enzyme that facilitates the transfer of a methyl group from S-adenosyl-L-methionine to glycine, resulting in the formation of S-adenosyl-L-homocysteine and sarcosine. This enzyme, found in the cytoplasm as a homotetramer, plays a crucial role in regulating cellular methylation processes. Mutations in the GNMT gene can lead to GNMT deficiency, also known as hypermethioninemia. GNMT influences DNA methylation by controlling the balance between S-adenosylmethionine and S-adenosylhomocysteine, and it participates in detoxification processes within liver cells. Notably, GNMT expression is reduced in human hepatocellular carcinoma (HCC).
Description
Recombinant human GNMT, with a 20-amino acid Histidine tag attached to its N-terminus, is produced in E. coli. This protein is a single, non-glycosylated polypeptide chain consisting of 315 amino acids (with amino acids 1-295 originating from the GNMT sequence) and has a molecular weight of 34.9 kDa. The purification of GNMT is achieved using proprietary chromatographic methods.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The GNMT solution is formulated in a buffer containing 20mM Tris (pH 8.0) and 20% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the GNMT solution should be stored at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To further enhance stability during long-term storage, adding a carrier protein like HSA or BSA (0.1%) is advisable. Repeated freezing and thawing of the solution should be avoided.
Purity
The purity of GNMT is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms
Glycine N-methyltransferase, GNMT.
Source
Escherichia Coli.
Amino Acid Sequence

MGSSHHHHHH SSGLVPRGSH MVDSVYRTRS LGVAAEGLPD QYADGEAARV WQLYIGDTRS RTAEYKAWLL GLLRQHGCQR VLDVACGTGV DSIMLVEEGF SVTSVDASDK MLKYALKERW NRRHEPAFDK WVIEEANWMT LDKDVPQSAE GGFDAVICLG NSFAHLPDCK GDQSEHRLAL KNIASMVRAG GLLVIDHRNY DHILSTGCAP PGKNIYYKSD LTKDVTTSVL IVNNKAHMVT LDYTVQVPGA GQDGSPGLSK FRLSYYPHCL ASFTELLQAA FGGKCQHSVL GDFKPYKPGQ TYIPCYFIHV LKRTD.

Q&A

What is the primary function of GNMT in human metabolism?

GNMT (Glycine N-Methyltransferase) is an enzyme that plays a crucial role in methionine metabolism, constituting approximately 1-3% of all cytosolic hepatic proteins . It catalyzes the conversion of glycine and S-adenosylmethionine (AdoMet or SAMe) to N-methylglycine and S-adenosylhomocysteine (AdoHcy or SAH) .

This reaction serves two critical functions: first, it contributes significantly to the breakdown of methionine; second, it helps maintain the optimal ratio of SAMe to SAH in cells, which is essential for proper methylation processes throughout the body . These methylation reactions are fundamental to numerous cellular functions, including DNA regulation, protein and lipid reactions, and neurotransmitter processing .

Beyond methylation control, GNMT also participates in the detoxification of xenobiotics in the liver, highlighting its multifunctional role in human metabolism .

How is GNMT deficiency diagnosed in clinical settings?

Diagnosing GNMT deficiency follows a multistep process. Initially, clinicians conduct a thorough medical history review, physical examination, and standard laboratory tests. Specialized blood and urine analyses prove particularly valuable in identifying characteristic biochemical abnormalities .

The definitive diagnosis requires molecular genetic testing to identify mutations in the GNMT gene . While newborn screening for this condition is available in the United States, GNMT deficiency is not currently included in standard newborn screening panels in many countries .

Key diagnostic markers include:

  • Elevated methionine levels in plasma

  • Increased S-adenosylmethionine (SAMe) concentrations

  • Normal homocysteine levels

  • Elevated serum transaminases

  • Mild hepatomegaly detected through imaging

It's worth noting that GNMT deficiency is extremely rare, with documentation of only five individuals from four different families to date .

Which GNMT polymorphisms have been identified, and how do they affect enzyme function?

Research has identified several significant polymorphisms in the GNMT gene that impact enzyme function. Three particularly well-documented polymorphisms include L49P, N140S, and H176N . These variants have been confirmed to decrease GNMT enzymatic activity, leading to clinical manifestations related to impaired methionine metabolism .

Interestingly, the structural basis for the detrimental effects of L49P and N140S substitutions can be predicted from crystal structures, while the destabilization caused by H176N is more difficult to discern through static structural analysis alone .

These polymorphisms constitute an important area for molecular dynamics studies to better understand how these subtle changes affect protein structure, dynamics, and enzymatic activity.

What is the prevalence of GNMT mutations and their associated inheritance patterns?

GNMT deficiency resulting from mutations in the GNMT gene is extraordinarily rare. According to current clinical data, only five individuals from four different families have been diagnosed with this condition worldwide . This extreme rarity makes it challenging to establish precise prevalence rates across different populations.

The condition follows an autosomal recessive inheritance pattern, requiring two copies of the mutated gene (one from each parent) for the disease to manifest. Carriers with only one copy of the mutation typically do not exhibit symptoms.

At least six distinct variants in the GNMT gene have been documented in individuals with hypermethioninemia, which is characterized by excess methionine in the blood . Most of these variants involve amino acid substitutions that reduce enzyme function. The resulting decrease in glycine N-methyltransferase activity impairs methionine breakdown, causing its accumulation in the bloodstream .

How does GNMT deficiency contribute to liver pathology based on knockout mouse models?

Studies using GNMT-knockout (GNMT-KO) mice have provided compelling evidence for GNMT's role in liver health. These animal models effectively recapitulate the biochemical abnormalities observed in humans with GNMT mutations .

GNMT-KO mice develop a progressive liver pathology characterized by:

  • Elevated serum aminotransferases (ALT and AST), indicating hepatocellular damage

  • Dramatically increased methionine and SAMe levels

  • Steatosis (fatty liver)

  • Progressive fibrosis

  • Development of hepatocellular carcinoma (HCC) with age

The biochemical basis for this pathology stems from profound methylation disturbances. In GNMT-KO mice, liver SAMe content is elevated 35-fold, and the critical SAMe/SAH ratio increases approximately 100-fold . This severe imbalance leads to aberrant methylation of DNA and histones, resulting in epigenetic modulation of carcinogenic pathways .

These findings highlight GNMT's essential role as a regulatory enzyme that removes excess SAMe and maintains homeostatic methylation balance in hepatic tissue. When this function is lost, the resulting methylation dysregulation contributes to progressive liver damage and eventual carcinogenesis.

What is the connection between GNMT expression and hepatocellular carcinoma (HCC)?

Multiple lines of evidence suggest a significant relationship between GNMT and hepatocellular carcinoma:

  • GNMT is notably absent in hepatocellular carcinoma tissues

  • GNMT expression is downregulated in the livers of patients at risk of developing HCC, including those with hepatitis C virus-induced and alcohol-induced cirrhosis

  • Loss of heterozygosity of the GNMT gene has been reported in approximately 40% of HCC patients

  • GNMT has been proposed as a tumor-susceptibility gene for liver cancer based on genetic studies

The mechanistic link between GNMT deficiency and carcinogenesis appears to involve epigenetic dysregulation. In GNMT-KO mice, researchers observed activation of the Ras and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathways, which are known to contribute to oncogenesis .

Additionally, alterations in DNA methylation patterns and histone H3K27 methylation were detected, suggesting that GNMT deficiency leads to epigenetic modulation of critical carcinogenic pathways . These epigenetic changes likely contribute to the progressive development of HCC in the context of GNMT deficiency.

What experimental approaches are most effective for studying GNMT structure-function relationships?

Investigating GNMT structure-function relationships requires a multifaceted approach combining several complementary methodologies:

  • X-ray Crystallography: Several crystal structures of rat and human GNMT homotetramers are available, including structures of variant forms like H176N . These provide static snapshots of protein structure that can reveal how polymorphisms might affect enzyme function.

  • Molecular Dynamics (MD) Simulations: These computational approaches overcome limitations of static crystal structures by providing high-resolution information on protein dynamics in solution. MD simulations are particularly valuable for polymorphic variants that may be difficult to crystallize due to instability or aggregation .

  • Enzymatic Activity Assays: Biochemical assays measuring the conversion of SAMe and glycine to SAH and sarcosine provide direct functional assessment of wild-type and variant GNMT proteins.

  • Site-Directed Mutagenesis: This technique allows researchers to introduce specific amino acid substitutions to study their effects on enzyme structure and function, helping to identify critical residues.

  • Animal Models: GNMT-knockout mice have proven invaluable for understanding the physiological consequences of GNMT deficiency . These models recapitulate many aspects of human GNMT deficiency and provide insights into disease progression.

For variants like H176N, where crystal structures fail to clearly explain functional deficits, combining structural data with MD simulations and biochemical characterization is particularly effective. This integrated approach can reveal how subtle changes in protein dynamics and conformational flexibility affect catalytic activity.

What considerations should researchers take when designing experiments to study GNMT methylation effects?

When investigating GNMT's role in regulating methylation processes, researchers should consider several critical experimental design factors:

  • SAMe/SAH Ratio Measurement: Since GNMT's primary function is maintaining the cellular SAMe/SAH ratio, accurate quantification of these metabolites is essential. Liquid chromatography-mass spectrometry (LC-MS) techniques provide the most reliable measurements.

  • Tissue Specificity: GNMT is expressed predominantly in the liver, pancreas, and prostate . Experimental designs should account for this tissue-specific expression pattern, particularly when using cell culture models.

  • Global vs. Locus-Specific Methylation Analysis: GNMT deficiency affects both global DNA methylation patterns and specific loci associated with carcinogenesis. Experimental designs should incorporate both broad methylation assays (e.g., HPLC-based global methylation analysis) and targeted approaches (e.g., bisulfite sequencing of specific regulatory regions).

  • Histone Methylation Assessment: Beyond DNA methylation, GNMT deficiency affects histone methylation patterns, particularly H3K27 . ChIP-seq approaches can identify genome-wide changes in histone modification patterns.

  • Temporal Considerations: In knockout models, methylation abnormalities precede visible pathology. Time-course experiments are essential to establish causality between methylation changes and disease progression.

When experimenting with cells or animals with altered GNMT expression, researchers should monitor not only direct methylation changes but also downstream effects on gene expression and signaling pathways, particularly those associated with carcinogenesis like Ras and JAK/STAT pathways .

What are the clinical manifestations of GNMT deficiency in humans compared to animal models?

The clinical presentation of GNMT deficiency shows both similarities and differences between humans and animal models:

Human Manifestations:

  • Mild hepatomegaly (liver enlargement)

  • Chronic elevation of serum transaminases

  • Hypermethioninemia (elevated methionine in blood)

  • Some individuals may be asymptomatic

  • Some cases report upper respiratory tract infections, failure to thrive, and febrile convulsions, though the relationship to GNMT deficiency remains uncertain

GNMT-Knockout Mouse Model Manifestations:

  • Elevated serum aminotransferases

  • Dramatically increased methionine and SAMe levels

  • Progressive development of steatosis (fatty liver)

  • Fibrosis of liver tissue

  • Development of hepatocellular carcinoma with age

The mouse model demonstrates a more severe progression of liver pathology than typically observed in human cases. This difference might reflect earlier detection in humans, genetic background effects, or environmental factors that modify disease expression.

The table below summarizes key biochemical findings in 8-month-old GNMT-KO mice compared to wild-type controls:

ParameterWild-type (8 months)GNMT-KO (8 months)
Methionine (μmol/L)51.6 ± 7.0Significantly elevated*

*Note: The exact value for GNMT-KO mice at 8 months is not fully provided in the search results, but is described as significantly elevated compared to wild-type .

How do researchers differentiate GNMT deficiency from other causes of hypermethioninemia?

Differentiating GNMT deficiency from other causes of hypermethioninemia requires a systematic diagnostic approach:

  • Biochemical Profile Analysis: GNMT deficiency presents with elevated methionine and SAMe levels but normal homocysteine concentrations . This pattern differs from other causes of hypermethioninemia:

    • Methionine adenosyltransferase deficiency: Low SAMe levels with elevated methionine

    • Cystathionine β-synthase deficiency: Elevated homocysteine with elevated methionine

  • Liver Function Assessment: GNMT deficiency typically shows mild to moderate liver dysfunction with elevated transaminases . The presence of hepatomegaly with otherwise minimal liver dysfunction is characteristic.

  • Genetic Testing: Definitive diagnosis requires identification of pathogenic variants in the GNMT gene through molecular genetic testing . This distinguishes GNMT deficiency from other genetic causes of hypermethioninemia.

  • Enzyme Activity Measurement: In specialized laboratories, direct measurement of GNMT enzyme activity in liver biopsy samples can confirm the diagnosis, though this is rarely necessary with modern genetic testing capabilities.

  • Exclusion of Secondary Causes: Researchers must rule out non-genetic causes of hypermethioninemia, including high protein intake, liver disease from other etiologies, and certain medications.

When conducting research on hypermethioninemia, comprehensive metabolic profiling of the methionine cycle intermediates, combined with genetic analysis, provides the most accurate differential diagnosis between GNMT deficiency and other disorders of methionine metabolism.

What are promising therapeutic approaches being investigated for GNMT deficiency?

While no established treatments exist specifically for GNMT deficiency, several therapeutic approaches deserve investigation:

  • Dietary Methionine Restriction: Since GNMT deficiency results in methionine and SAMe accumulation, restricting dietary methionine intake may help normalize these metabolites and prevent downstream pathology. This approach requires careful nutritional monitoring, particularly in growing children.

  • SAMe-Lowering Agents: Compounds that facilitate alternative pathways for SAMe utilization or metabolism could potentially reduce the SAMe/SAH imbalance characteristic of GNMT deficiency.

  • Gene Therapy Approaches: Given that GNMT deficiency results from specific genetic mutations, gene replacement or gene editing strategies using CRISPR-Cas9 or similar technologies represent promising future directions, particularly for liver-targeted delivery.

  • Epigenetic Modifiers: Since many pathological consequences of GNMT deficiency stem from aberrant methylation patterns, epigenetic modifying drugs that can normalize these patterns may offer therapeutic benefit.

  • Antioxidant Therapies: GNMT deficiency may increase oxidative stress in hepatic tissue. Targeted antioxidant therapies could potentially mitigate this component of liver injury.

Research into these approaches is still in early stages, and translational studies are needed to determine efficacy and safety. The development of improved animal models that more closely recapitulate human GNMT deficiency would accelerate therapeutic discovery.

What research gaps remain in understanding the role of GNMT in methylation regulation and disease pathogenesis?

Despite significant advances in GNMT research, several critical knowledge gaps remain:

  • Tissue-Specific Effects: While GNMT's role in liver has been extensively studied, its function in other tissues where it is expressed (pancreas, prostate) remains poorly characterized . Further research is needed to understand tissue-specific consequences of GNMT deficiency.

  • Interaction with Environmental Factors: How environmental exposures, including diet, toxins, and medications, interact with GNMT deficiency to modify disease expression remains largely unknown.

  • Temporal Dynamics of Epigenetic Changes: The precise sequence of epigenetic alterations following GNMT deficiency and how these changes progress to carcinogenesis require further elucidation through time-course studies.

  • Genotype-Phenotype Correlations: With only five documented human cases, the relationship between specific GNMT mutations and clinical phenotypes remains unclear . Additional case reports and functional studies of different mutations would help establish these correlations.

  • Polymorphic Variants in Population Health: The significance of common GNMT polymorphisms in population health and disease susceptibility beyond identified cases of complete deficiency represents an important area for epidemiological research.

  • Compensatory Mechanisms: Identifying potential compensatory pathways that may activate in GNMT deficiency could reveal new therapeutic targets and explain phenotypic variability.

Addressing these research gaps will require multidisciplinary approaches combining molecular biology, structural biochemistry, epigenetics, and clinical research to fully understand GNMT's role in human health and disease.

What are the best approaches for measuring GNMT activity in different biological samples?

Accurate measurement of GNMT activity requires specialized techniques tailored to different sample types:

  • Liver Tissue Samples:

    • Radiometric assay using [methyl-³H]SAMe as methyl donor and glycine as acceptor, measuring the formation of radiolabeled sarcosine

    • HPLC-based methods measuring the conversion of SAMe to SAH

    • Mass spectrometry approaches for high-sensitivity detection of reaction products

  • Cell Culture Models:

    • Stable isotope tracing using ¹³C-labeled methionine followed by metabolite analysis via LC-MS/MS

    • In situ activity assays in permeabilized cells to maintain cellular context

    • Immunoprecipitation of GNMT followed by activity measurement to assess specific activity

  • Blood Samples:

    • Direct enzyme activity measurement is challenging in blood

    • Surrogate markers including methionine and SAMe levels provide indirect assessment

    • Genotyping for known functional variants may predict activity levels

For all sample types, appropriate controls are essential, including:

  • Positive controls with recombinant GNMT protein

  • Negative controls with GNMT-specific inhibitors

  • Samples with known GNMT deficiency (when available)

Temperature, pH, and substrate concentrations should be carefully standardized, as these factors significantly influence GNMT activity measurements. Additionally, researchers should be aware that post-translational modifications may affect enzyme activity, necessitating careful sample handling to preserve in vivo activity states.

How can researchers effectively model GNMT deficiency in experimental systems?

Creating accurate experimental models of GNMT deficiency requires careful consideration of several approaches:

  • Animal Models:

    • GNMT-knockout mice provide the most comprehensive in vivo model, recapitulating key biochemical and pathological features of human deficiency

    • Conditional knockout models can help distinguish developmental versus adult-onset effects

    • Humanized mouse models expressing human GNMT variants may better reflect human polymorphisms

  • Cell Culture Systems:

    • CRISPR-Cas9 engineered GNMT-knockout hepatocyte lines

    • siRNA or shRNA-mediated GNMT knockdown for transient suppression

    • Introduction of specific human GNMT variants through plasmid transfection

    • iPSC-derived hepatocytes from patients with GNMT deficiency (if available)

  • Organoid Models:

    • Liver organoids with GNMT mutations better recapitulate the three-dimensional tissue architecture

    • Co-culture systems incorporating multiple cell types can model tissue interactions

  • Computational Models:

    • Molecular dynamics simulations of wild-type and variant GNMT proteins

    • Metabolic flux analysis to predict consequences of GNMT deficiency on related pathways

    • Systems biology approaches integrating transcriptomic, proteomic, and metabolomic data

When designing these models, researchers should consider:

  • The degree of GNMT suppression (partial vs. complete)

  • Acute versus chronic deficiency effects

  • Developmental timing of GNMT deficiency

  • Genetic background effects that may modify phenotypes

Validation of these models should include confirmation of altered SAMe/SAH ratios, methylation status assessment, and comparison to known human phenotypes where possible.

How might GNMT function in tissue types beyond the liver, and what are the implications for disease?

While GNMT is predominantly studied in liver tissue, emerging research indicates important functions in other tissues:

  • Pancreatic Tissue:

    • GNMT is expressed in pancreatic tissue, suggesting a role in pancreatic methionine metabolism

    • Potential implications for pancreatic disorders, including pancreatitis and pancreatic cancer

    • Research needed to determine if GNMT deficiency affects pancreatic methylation status and function

  • Prostate Tissue:

    • GNMT expression in prostate tissue suggests relevance to prostate biology

    • Certain GNMT gene variations have been associated with increased risk of prostate cancer

    • Further investigation needed to understand the mechanistic link between GNMT function and prostate pathology

  • Neural Tissue:

    • Although not traditionally considered a major site of GNMT expression, the neurological symptoms occasionally reported in hypermethioninemia suggest potential neurological implications

    • Research needed to determine if GNMT plays any role in neural methylation processes

    • Possible indirect effects through altered systemic methionine metabolism

  • Immune System:

    • Methylation processes regulate immune cell function and differentiation

    • The role of GNMT in immune tissues remains largely unexplored

    • Potential implications for autoimmune conditions and inflammatory responses

Understanding GNMT's function in these non-hepatic tissues requires:

  • Tissue-specific knockout models

  • Cell-type specific expression analysis

  • Methylome and transcriptome profiling of affected tissues

  • Clinical correlations between GNMT variants and non-hepatic disease manifestations

This represents an important frontier in GNMT research with potential implications for understanding the full spectrum of GNMT-related pathology.

What is the relationship between GNMT function and other methyltransferases in the regulation of cellular methylation?

GNMT functions within a complex network of methyltransferases that collectively regulate cellular methylation status:

  • Competitive SAMe Utilization:

    • GNMT uses approximately 1% of total SAMe in hepatic tissue

    • Competes with DNA methyltransferases (DNMTs), histone methyltransferases (HMTs), and other methyltransferases for the methyl donor SAMe

    • Serves as a regulatory "sink" for excess SAMe, preventing aberrant methylation by other enzymes

  • Hierarchical Enzyme Kinetics:

    • Different methyltransferases have varying affinities for SAMe and sensitivities to SAH inhibition

    • This creates a hierarchical response to changing SAMe/SAH ratios

    • GNMT deficiency disrupts this hierarchy, leading to preferential activity of certain methyltransferases

  • Feedback Regulation:

    • Complex regulatory mechanisms exist between different methyltransferases

    • Changes in GNMT activity can indirectly affect the activity of other methyltransferases through altered substrate availability and product inhibition

    • Research needed to map these regulatory networks comprehensively

  • Tissue-Specific Interactions:

    • The relative importance of GNMT versus other methyltransferases varies by tissue

    • In liver, GNMT plays a dominant role in SAMe homeostasis

    • In tissues with lower GNMT expression, other regulatory mechanisms may predominate

  • Pathological Consequences of Imbalance:

    • GNMT deficiency leads to aberrant DNA and histone methylation

    • Research needed to determine which specific methyltransferases are most affected by GNMT deficiency

    • Therapeutic approaches might target specific downstream methyltransferases rather than GNMT itself

Understanding these complex interactions requires systems biology approaches integrating:

  • Enzyme kinetics data

  • Metabolic flux analysis

  • Tissue-specific expression patterns

  • Mathematical modeling of the methylation network

Product Science Overview

Introduction

Glycine N-methyltransferase (GNMT) is an enzyme that plays a crucial role in the metabolism of methyl groups in the human body. This enzyme is encoded by the GNMT gene and is primarily found in the liver, but it is also present in other tissues such as the pancreas and kidneys .

Structure and Function

GNMT is a tetrameric cytosolic protein that catalyzes the transfer of a methyl group from S-adenosylmethionine (AdoMet) to glycine, producing S-adenosylhomocysteine (AdoHcy) and sarcosine . This reaction is essential for maintaining normal levels of AdoMet, which is a critical methyl donor in various biological processes .

The enzyme’s structure allows it to bind to both AdoMet and glycine, facilitating the methylation process. The human recombinant form of GNMT is produced using E. coli expression systems, which ensures high purity and activity .

Biological Significance

GNMT plays a significant role in the regulation of methyl group metabolism. By converting glycine to sarcosine, it helps maintain the balance of AdoMet and AdoHcy, which is vital for numerous methylation reactions in the body . These reactions are involved in DNA methylation, protein methylation, and the synthesis of various biomolecules.

In the liver, GNMT is particularly abundant, constituting up to 3% of the soluble protein in liver cytosol . Its activity is also observed in other tissues, including the pancreas, kidneys, and certain regions of the brain .

Clinical Relevance

Mutations in the GNMT gene can lead to various metabolic disorders. For instance, deficiencies in GNMT activity have been linked to hypermethioninemia, a condition characterized by elevated levels of methionine in the blood . Additionally, GNMT has been implicated in liver diseases, including hepatocellular carcinoma, where its expression is often altered .

Applications of Human Recombinant GNMT

The recombinant form of GNMT is used in various research applications to study its function and role in metabolism. It is also employed in drug development and biochemical assays to investigate potential therapeutic targets for metabolic and liver diseases .

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.