The MAT1A gene produces two isoforms of methionine adenosyltransferase:
MATI: A homotetramer (four identical subunits)
MATIII: A homodimer (two subunits)
Both convert methionine to S-adenosylmethionine (SAMe), the primary donor of methyl groups for DNA methylation, lipid synthesis, and neurotransmitter regulation .
Methionine + ATP → SAMe + Tripolyphosphate
Tripolyphosphate → PPi + Pi
SAMe is essential for:
DNA methylation (epigenetic regulation)
Polyamine synthesis
Over 70 MAT1A mutations cause hypermethioninemia, characterized by elevated plasma methionine and reduced SAMe levels .
Mutation Type | Effect | Outcome |
---|---|---|
Missense substitutions | Altered enzyme activity | Reduced SAMe production . |
Premature stop codons | Truncated, nonfunctional enzyme | Accumulated methionine . |
Nonalcoholic Steatohepatitis (NASH): MAT1A-knockout mice develop hepatic steatosis and inflammation .
Hepatocellular Carcinoma (HCC): MAT1A silencing correlates with dedifferentiation and tumor growth .
In non-small cell lung cancer (NSCLC), MAT1A stabilizes CCND1 (cyclin D1) by inhibiting SKP2-mediated ubiquitination, promoting glycolysis and tumorigenesis .
CCND1 Stabilization: MAT1A binds SKP2, reducing CCND1 degradation .
Glycolysis Activation: Upregulates glycolytic enzymes (e.g., GLUT1, HK2) .
Antisense oligonucleotides (ASOs) targeting Mat1a in obese mice:
The MAT1A promoter contains binding sites for:
C/EBP (CAAT/enhancer-binding protein)
HNF (hepatocyte nuclear factor)
Deletions in regions -705 to -839 bp and -1111 to -1483 bp alter promoter activity in liver cells .
Factor | Role |
---|---|
C/EBP | Enhances MAT1A expression in liver cells . |
Glucocorticoids | Induce MAT1A expression via promoter activation . |
The MAT1A gene provides instructions for producing methionine adenosyltransferase, an essential enzyme in methionine metabolism. This enzyme is expressed in two oligomeric forms: MATI (a homotetramer of four identical protein subunits) and MATIII (a homodimer of two identical protein subunits). Both forms are predominantly found in the liver, though MAT1A expression has also been detected in pancreas and, in smaller amounts, in most tissues including brain .
The primary function of methionine adenosyltransferase is catalyzing the conversion of methionine and ATP to S-adenosylmethionine (AdoMet or SAMe). This enzymatic reaction is critical for transmethylation, a process involving the transfer of methyl groups (consisting of one carbon atom and three hydrogen atoms) to various compounds. Transmethylation plays vital roles in numerous cellular processes, including:
Regulation of gene expression through DNA methylation
Control of protein and lipid interactions
This enzyme has been remarkably conserved throughout evolution, with perfect conservation of active site residues across 292 MAT genes studied in bacteria and eukaryotes .
Humans possess two genes encoding methionine adenosyltransferase isoforms:
MAT1A - Encodes the catalytic subunit that forms MAT I (tetrameric) and MAT III (dimeric) holoenzymes
MAT2A - Encodes the catalytic subunit of MAT II
Evolutionary analysis suggests these genes resulted from a duplication event that occurred after the human lineage diverged from sea squirts (Urochordata) but before divergence from jawed vertebrates (Teleostomi), approximately 400-500 million years ago. The two human MAT genes encode amino acid sequences with approximately 85% identity .
Methodological approaches for studying MAT1A include:
Gene expression analysis:
qRT-PCR for quantifying MAT1A mRNA levels
RNA sequencing for comprehensive transcriptomic profiling
In situ hybridization for localization studies
Protein analysis:
Western blotting with MAT1A-specific antibodies
Immunohistochemistry for tissue localization
Proteomic approaches to identify interaction partners
Enzymatic activity assays:
These methodologies enable comprehensive characterization of MAT1A expression patterns, helping researchers understand tissue-specific roles and developmental regulation of this critical enzyme.
Over 70 variants (mutations) in the MAT1A gene have been identified in individuals with hypermethioninemia. These mutations can be categorized into several types:
Missense mutations - These substitute one amino acid for another in the methionine adenosyltransferase enzyme, typically reducing enzymatic efficiency. Examples include R264H, R264C, and G336R .
Nonsense and truncating mutations - These introduce premature stop signals, resulting in shortened, nonfunctional enzymes .
Splicing mutations - These affect normal transcript processing, leading to abnormal protein products .
The impact on enzyme function varies considerably:
Complete loss-of-function mutations typically cause severe elevations in plasma methionine
Missense mutations often result in partial enzyme activity with moderate methionine elevation
The dominant R264H mutation uniquely renders dimers formed between mutant and wild-type subunits inactive
Effective characterization of novel MAT1A mutations involves a multi-step approach:
Genetic analysis:
Functional characterization:
For example, researchers have identified novel mutations by amplifying each MAT1A exon, analyzing products on mutation-detection-enhancement nondenaturing gels, and confirming mutations by sequencing at least five subclones of each .
The genotype-phenotype relationship in MAT1A deficiency shows considerable variability:
Genotype Category | Typical Methionine Levels | Clinical Manifestations |
---|---|---|
Compound heterozygote for truncating and severely inactivating missense mutations | 1,226–1,870 μM (normal: 5-35 μM) | Brain gray matter abnormalities or demyelination |
Compound heterozygote for truncating and less severe inactivating mutations | 770–1,240 μM | Mild cognitive impairment |
Two inactivating missense mutations or heterozygous R264H | 105–467 μM | Typically clinically unaffected |
This complex relationship underscores the necessity for further studies to firmly establish correlations between genotypes and clinical phenotypes in MAT I/III deficiency .
MAT1A deficiency presents with remarkable clinical heterogeneity:
Asymptomatic hypermethioninemia:
Many individuals have elevated methionine levels without clinical manifestations
Often detected through newborn screening programs designed to identify homocystinuria
Methionine levels typically in the 100-500 μM range (normal: 5-35 μM)
Symptomatic MAT1A deficiency:
Neurological manifestations ranging from mild cognitive impairment to significant abnormalities
Brain demyelination observed in some cases
Gray matter abnormalities reported in others
Typically associated with higher methionine levels (>1000 μM)
A comprehensive survey of 64 patients with MAT1A mutations found exactly half (32 patients) had evidence of CNS abnormalities, highlighting the variable penetrance of neurological manifestations .
A systematic diagnostic approach includes:
Initial screening:
Elevated plasma methionine (>35 μM) without elevation of homocysteine or tyrosine
Exclusion of secondary causes of hypermethioninemia (tyrosinemia type I, cystathionine β-synthase deficiency, liver disease)
Confirmatory testing:
Sequencing of the MAT1A gene to identify mutations
Assessment of mutation type and zygosity
Special attention to the R264H mutation, which causes dominant hypermethioninemia
Clinical evaluation:
The pattern of early-onset, persistent, isolated hypermethioninemia without elevation of homocysteine or tyrosine is characteristic of MAT I/III deficiency .
Distinguishing between benign and potentially pathological forms requires:
Genetic analysis:
Heterozygous R264H mutation typically causes benign dominant hypermethioninemia
Homozygous or compound heterozygous mutations, particularly those severely affecting enzyme function, may pose greater risk for neurological complications
Biochemical severity:
Methionine levels >1000 μM associated with increased risk of neurological manifestations
Moderate elevations (100-500 μM) more often associated with benign outcomes
Longitudinal monitoring:
Research suggests that while plasma methionine level correlates somewhat with clinical severity, the relationship between genotype and neurological outcome remains complex and incompletely understood .
While not explicitly detailed in the search results, several experimental approaches can advance understanding of MAT1A deficiency:
Cellular models:
Primary hepatocytes with MAT1A knockdown or knockout
Induced pluripotent stem cells (iPSCs) derived from patient samples
Cell lines expressing specific MAT1A mutations
Animal models:
MAT1A knockout mice to study systemic effects of enzyme deficiency
Conditional and tissue-specific knockouts to examine MAT1A's role in specific organs
Knock-in models of human mutations to study genotype-phenotype correlations
Ex vivo systems:
Liver slices or organoids for studying hepatic methionine metabolism
Brain tissue models for investigating effects on myelination
These experimental systems can help elucidate mechanisms of disease, identify potential biomarkers, and test therapeutic interventions.
Neuroimaging plays a crucial role in characterizing MAT1A-associated CNS abnormalities:
Magnetic Resonance Imaging (MRI):
T2-weighted and FLAIR sequences to detect white matter abnormalities
Diffusion tensor imaging (DTI) to assess white matter tract integrity
Spectroscopy to evaluate metabolite profiles in affected brain regions
Longitudinal imaging protocols:
Serial MRIs to monitor disease progression or treatment response
Standardized acquisition parameters for consistent evaluation
Quantitative analysis of myelination patterns and brain volume
The search results mention MRI findings in several patients, indicating this modality's importance in clinical evaluation. Normal myelination was reported in some patients with mild symptoms, while others with more severe manifestations showed abnormal MRI findings .
Integrated multi-omics approaches offer powerful tools for comprehensive investigation:
Genomics and transcriptomics:
Whole genome/exome sequencing to identify novel variants
RNA-seq to examine tissue-specific expression patterns
Single-cell sequencing to understand cellular heterogeneity
Proteomics and metabolomics:
Quantification of MAT1A protein levels and post-translational modifications
Measurement of methionine cycle metabolites
Global metabolomic profiling to identify perturbed pathways
Epigenomics:
Genome-wide methylation analysis to assess consequences of AdoMet deficiency
Chromatin immunoprecipitation sequencing (ChIP-seq) to evaluate histone modifications
Investigation of non-coding RNA regulation
These approaches can reveal how MAT1A deficiency affects multiple biological systems and identify potential biomarkers and therapeutic targets.
While the search results mention "a variety of treatment regimens" without specific details, potential therapeutic approaches include:
Dietary interventions:
Methionine-restricted diet to reduce plasma methionine levels
Formulation of appropriate dietary recommendations based on mutation severity and clinical presentation
Supplementation strategies:
S-adenosylmethionine (AdoMet/SAMe) supplementation to bypass the metabolic block
Other methyl donors to support methylation reactions
Monitoring and preventive care:
One goal of the collaborative study described in the search results was "to evaluate the outcomes of the variety of treatment regimens," suggesting various therapeutic approaches have been attempted but their efficacy remains to be firmly established .
Designing rigorous clinical trials for MAT1A deficiency requires addressing several methodological challenges:
These methodological considerations are essential for generating robust evidence to guide clinical management of this rare disorder.
Emerging therapeutic strategies could include:
Gene therapy approaches:
Liver-directed gene therapy to restore functional MAT1A expression
mRNA therapeutics for transient expression of functional enzyme
Gene editing technologies to correct specific mutations
Enzyme replacement or enhancement:
Development of stabilized recombinant MAT enzyme formulations
Small molecule chaperones to enhance residual enzyme activity
Novel delivery systems for targeting the liver
Metabolic bypass strategies:
Optimization of AdoMet supplementation regimens
Development of novel methyl donors with enhanced bioavailability
Targeted interventions for downstream methylation-dependent processes
While these approaches remain theoretical for MAT1A deficiency, they represent promising directions for future research based on advances in related metabolic disorders.
The MAT1A gene is located on chromosome 10q22.3 and encodes the alpha subunit of the enzyme. The protein can exist in different forms, such as a homotetramer (MAT I) or a homodimer (MAT III), depending on the cellular context . The recombinant form of this protein is often produced in various expression systems, including HEK293T cells and Nicotiana tabacum (tobacco) plants .
MAT1A catalyzes a two-step reaction:
SAMe is a vital molecule that serves as a methyl group donor in numerous methylation reactions, which are essential for the regulation of gene expression, protein function, and lipid metabolism .
Mutations in the MAT1A gene can lead to methionine adenosyltransferase deficiency, a rare metabolic disorder characterized by elevated levels of methionine in the blood (hypermethioninemia). This condition can result in various clinical manifestations, including neurological symptoms and liver dysfunction .
The recombinant MAT1A protein is typically produced using advanced expression systems. For example, the ALiCE® system, based on a lysate obtained from Nicotiana tabacum, is used to produce even the most challenging proteins. The protein is then purified through multi-step processes to ensure correct folding and modification .