PSAT1 Human

Phosphoserine Aminotransferase 1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

PSAT1 Human Recombinant produced in E.coli is a single, non-glycosylated polypeptide chain containing 394 amino acids (1-370) and having a molecular mass of 42.9kDa.
PSAT1 is fused to a 24 amino acid His-tag at N-terminus & purified by proprietary chromatographic techniques.

Product Specs

Introduction
Phosphoserine aminotransferase (PSAT1) is an enzyme that plays a crucial role in the biosynthesis of serine, an amino acid essential for various biological processes. PSAT1 catalyzes the conversion of 3-phosphohydroxypyruvate to 3-phosphoserine, which is then dephosphorylated to form L-serine. This enzyme exhibits high expression levels in vital organs such as the brain, liver, kidney, and pancreas, while its expression is limited in tissues like the thymus, prostate, testis, and colon. Genetic defects in the PSAT1 gene can lead to phosphoserine aminotransferase deficiency (PSATD), a disorder characterized by low serine and glycine levels in plasma and cerebrospinal fluid. PSATD manifests clinically as intractable seizures, acquired microcephaly, hypertonia, and psychomotor retardation.
Description
This product consists of the recombinant human PSAT1 protein, expressed in E. coli and purified to a high degree. The protein is a single, non-glycosylated polypeptide chain comprising 394 amino acids (residues 1-370) with a molecular weight of 42.9 kDa. A 24 amino acid His-tag is fused to the N-terminus to facilitate purification by proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The PSAT1 protein is supplied in a solution at a concentration of 1 mg/ml. The storage buffer is 20mM Tris-HCl (pH 8.0) containing 20% glycerol and 1mM DTT.
Stability
For short-term storage (up to 4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. To ensure long-term stability, the addition of a carrier protein (0.1% HSA or BSA) is recommended. Repeated freezing and thawing should be avoided.
Purity
The purity of the PSAT1 protein is determined to be greater than 90.0% by SDS-PAGE analysis.
Synonyms
Phosphoserine aminotransferase, Phosphohydroxythreonine aminotransferase, PSAT, PSAT1, PSA, EPIP.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMDAPRQ VVNFGPGPAK LPHSVLLEIQ KELLDYKGVG ISVLEMSHRS SDFAKIINNT ENLVRELLAV PDNYKVIFLQ GGGCGQFSAV PLNLIGLKAG RCADYVVTGA WSAKAAEEAK KFGTINIVHP KLGSYTKIPD PSTWNLNPDA SYVYYCANET VHGVEFDFIP DVKGAVLVCD MSSNFLSKPV DVSKFGVIFA GAQKNVGSAG VTVVIVRDDL LGFALRECPS VLEYKVQAGN SSLYNTPPCF SIYVMGLVLE WIKNNGGAAA MEKLSSIKSQ TIYEIIDNSQ GFYVCPVEPQ NRSKMNIPFR IGNAKGDDAL EKRFLDKALE LNMLSLKGHR SVGGIRASLY NAVTIEDVQK LAAFMKKFLE MHQL.

Q&A

What is the molecular structure of human PSAT1 and how does it affect its function?

Human PSAT1 is a pyridoxal-phosphate (PLP)-dependent cytosolic protein that forms an active α/β domain structure homodimer. Each monomer is approximately 40 kDa and consists of 370 amino acids . The enzyme contains a catalytic lysine residue and bound PLP in the active site, which is essential for its aminotransferase activity . The dimeric structure is critical for proper enzymatic function, as demonstrated by SDS-PAGE analysis showing a 40 kDa band under reducing conditions .

The structural organization of PSAT1 directly influences its catalytic mechanism. The enzyme's active site accommodates both 3-phosphohydroxypyruvate and α-ketoglutarate as substrates, enabling the reversible second step in the de novo serine synthesis pathway. Understanding this structure-function relationship is fundamental for designing experiments to investigate PSAT1 activity and for developing potential inhibitors for therapeutic purposes.

How is PSAT1 activity measured in experimental settings?

PSAT1 enzymatic activity can be measured through spectrophotometric assays that monitor the conversion of NADH to NAD+ during the coupled reaction. A standard protocol involves:

  • Preparing a substrate mixture containing 0.4 mM NADH, 2 mM α-ketoglutaric acid, 40 μM pyridoxal 5'-phosphate, 2 mM O-phospho-L-serine, and recombinant PHGDH (as a coupling enzyme)

  • Adding the PSAT1 enzyme (typically at 20 μg/mL) to initiate the reaction

  • Monitoring the decrease in absorbance at 340 nm, which corresponds to NADH oxidation

  • Calculating specific activity using the formula:

Specific Activity (pmol/min/μg) =Adjusted Vmax (OD/min) × well volume (L) × 10¹² pmol/mol × (-1)
Extinction coefficient (M⁻¹cm⁻¹) × path correction (cm) × amount of enzyme (μg)

Using the extinction coefficient of 6220 M⁻¹cm⁻¹ for NADH and a path correction of 0.32 cm .

This methodology provides a quantitative measure of PSAT1 catalytic efficiency and can be used to evaluate the impact of mutations, inhibitors, or environmental conditions on enzyme function.

What are the tissue-specific expression patterns of PSAT1 and their physiological significance?

PSAT1 expression is most concentrated in the brain, liver, kidney, and pancreas, suggesting these tissues are primary sites for de novo serine synthesis . This expression pattern has significant physiological implications:

  • Brain expression is particularly critical because serine is considered essential for brain function due to insufficient delivery through the blood-brain barrier

  • Liver and kidney expression reflects their roles in amino acid metabolism and homeostasis

  • Pancreatic expression suggests involvement in metabolic regulation and potentially in cellular energy metabolism

The tissue-specific expression of PSAT1 correlates with the clinical manifestations observed in PSAT1-deficient patients, who often present with neurological symptoms including peripheral neuropathy, developmental delays, and seizures . Understanding these expression patterns is essential for designing tissue-specific experimental approaches and for interpreting the systemic effects of PSAT1 dysfunction.

What is the spectrum of phenotypes associated with PSAT1 mutations?

PSAT1-related disorders show remarkable clinical heterogeneity, ranging from severe Neu-Laxova syndrome to milder juvenile-onset neuropathy. The phenotypic spectrum includes:

  • Severe prenatal/neonatal presentation: Characterized by intrauterine growth restriction, microcephaly, ichthyosis, and early mortality (18 of 31 reported patients died at birth or within 1 month)

  • Childhood-onset neurodegenerative phenotype: Featuring developmental delays, seizures, and progressive neurological deterioration

  • Juvenile-onset peripheral neuropathy: A milder phenotype characterized by ichthyosis and progressive peripheral neuropathy without significant developmental delays

  • Adult-onset multiple system degeneration: Recently described phenotype including intrauterine growth restriction, childhood intellectual disability, adolescent gait abnormalities, and adult peripheral neuropathy with primary ovarian insufficiency

This spectrum demonstrates that juvenile-onset peripheral neuropathy might represent the mildest end of serine deficiency disorders . The phenotypic variability suggests that different mutations may affect PSAT1 activity to varying degrees, with potential genotype-phenotype correlations that warrant further investigation.

How do researchers differentiate between PSAT1-related disorders and other neurological conditions with similar presentations?

Differentiating PSAT1-related disorders from other neurological conditions requires a multi-faceted approach:

  • Clinical assessment: Evaluating the constellation of symptoms, including ichthyosis, peripheral neuropathy, developmental delays, and seizures

  • Biochemical testing: Measuring serine and glycine levels in plasma and cerebrospinal fluid, which are typically low in PSAT1 deficiency

  • Electrophysiological studies: Conducting nerve conduction studies to characterize the pattern and severity of peripheral neuropathy

  • Genetic analysis: Sequencing the PSAT1 gene to identify pathogenic variants, which is the definitive diagnostic approach

  • Exclusion of other etiologies: Investigating inflammatory, toxic, or other metabolic causes through appropriate laboratory tests

The methodological approach should be systematic, starting with clinical and biochemical assessments before proceeding to genetic testing. Researchers should be aware that milder phenotypes might be misdiagnosed as other forms of hereditary neuropathy, emphasizing the importance of considering PSAT1 deficiency in the differential diagnosis of juvenile-onset peripheral neuropathy with ichthyosis.

What is the recommended methodology for analyzing PSAT1 mutations in clinical samples?

The recommended methodology for analyzing PSAT1 mutations involves:

  • DNA extraction from peripheral blood or appropriate tissue samples

  • Targeted gene sequencing of PSAT1 or inclusion in gene panels for:

    • Serine synthesis disorders

    • Hereditary peripheral neuropathies

    • Ichthyosis-associated conditions

  • Whole exome sequencing as a more comprehensive approach when targeted testing is negative but clinical suspicion remains high

  • Validation of novel variants through segregation analysis in family members

  • Functional characterization of variants of uncertain significance through:

    • In vitro enzymatic activity assays using recombinant proteins

    • Structural modeling to predict the impact on protein folding and function

    • Cell-based assays to assess serine synthesis pathway activity

For mutations identified in research settings, it's crucial to establish their pathogenicity through population frequency data, in silico prediction tools, and functional studies. The c.296C>T mutation appears to be the most common pathogenic variant, accounting for 16.1% of reported alleles , which may serve as a positive control in validation studies.

How can PSAT1 activity be measured in human tissue samples for research purposes?

Measuring PSAT1 activity in human tissue samples requires specialized methodologies:

  • Tissue homogenization and subcellular fractionation:

    • Homogenize tissue samples in appropriate buffer containing protease inhibitors

    • Separate cytosolic fraction (where PSAT1 is primarily located) through differential centrifugation

  • Enzymatic activity assay:

    • Adapt the spectrophotometric NADH-coupled assay for tissue homogenates

    • Include appropriate controls to account for background activity from other enzymes

    • Normalize activity to protein concentration

  • ELISA-based quantification:

    • Use PSAT1-specific antibodies in an ELISA format to quantify protein levels

    • Follow standardized ELISA protocols using pre-coated microplates with PSAT1 antibodies

    • Establish standard curves using purified recombinant PSAT1 protein

  • Western blot analysis:

    • Use validated PSAT1 antibodies to detect expression levels

    • Compare with housekeeping proteins for normalization

    • Consider analyzing both monomeric (40 kDa) and potential dimeric forms

  • Correlation with serine metabolite levels:

    • Measure serine and related metabolites in the same samples

    • Correlate enzyme activity with metabolite concentrations to assess functional impact

These methodological approaches provide complementary information about PSAT1 expression and activity, allowing researchers to comprehensively characterize enzyme function in different tissues and disease states.

What are the latest methodologies for investigating PSAT1's role in cancer progression?

Recent methodologies for investigating PSAT1's role in cancer progression include:

  • Gene expression modulation techniques:

    • CRISPR-Cas9 gene editing to create PSAT1 knockout cancer cell lines

    • shRNA or siRNA for transient knockdown of PSAT1 expression

    • Overexpression systems to study gain-of-function effects

  • Metabolic flux analysis:

    • Isotope tracing with ¹³C-labeled glucose or glutamine to track serine synthesis

    • Mass spectrometry to quantify labeled metabolites

    • Computational modeling of metabolic pathways

  • Signaling pathway analysis:

    • Investigating PSAT1's interaction with the GSK3β/β-catenin pathway

    • Studying effects on α-ketoglutarate levels and downstream signaling

    • Examining non-canonical functions independent of serine synthesis

  • In vivo models:

    • Xenograft models with PSAT1-modified cancer cells

    • Patient-derived xenografts to study PSAT1 in a more clinically relevant context

    • Conditional knockout mouse models to study tissue-specific effects

  • Clinical correlation studies:

    • Analysis of PSAT1 expression in tumor samples and correlation with prognosis

    • Investigation of PSAT1 as a biomarker in nasopharyngeal, breast, and esophageal carcinomas

    • Assessment of PSAT1 as a therapeutic target in colorectal, esophageal, and breast cancers

These methodologies collectively provide a comprehensive approach to understanding PSAT1's multifaceted roles in cancer, from metabolic reprogramming to direct signaling effects that promote proliferation and metastasis.

How can researchers design experiments to distinguish between PSAT1's canonical and non-canonical functions?

Distinguishing between PSAT1's canonical role in serine synthesis and its emerging non-canonical functions requires carefully designed experimental approaches:

  • Substrate availability manipulation:

    • Culture cells in serine-depleted medium to emphasize the canonical pathway

    • Provide excess serine to potentially isolate non-canonical functions

    • Use isotope-labeled precursors to track metabolic flux through the canonical pathway

  • Enzyme activity-deficient mutants:

    • Generate catalytically inactive PSAT1 mutants (targeting the PLP binding site)

    • Compare phenotypic effects of wild-type versus catalytically inactive PSAT1

    • Assess whether non-canonical functions persist despite loss of enzymatic activity

  • Interactome analysis:

    • Perform immunoprecipitation followed by mass spectrometry to identify protein binding partners

    • Validate key interactions through co-immunoprecipitation and proximity ligation assays

    • Map interaction domains to identify regions involved in non-canonical functions

  • Domain-specific mutations:

    • Create truncation or point mutations targeting specific protein domains

    • Assess which domains are essential for canonical versus non-canonical functions

    • Use structural biology approaches to understand mechanism

  • Temporal dynamics analysis:

    • Study acute versus chronic PSAT1 depletion effects

    • Investigate the kinetics of metabolic versus signaling responses

    • Use inducible systems to control PSAT1 expression with temporal precision

These approaches can help researchers delineate PSAT1's roles in GSK3β/β-catenin signaling, α-ketoglutarate regulation for stem cell self-renewal, and migratory/proliferative functions independent of serine synthesis , advancing our understanding of this multifunctional enzyme.

What are the current approaches for developing PSAT1 inhibitors for potential therapeutic applications?

Developing PSAT1 inhibitors for therapeutic applications involves several strategic approaches:

  • Structure-based drug design:

    • Utilize crystal structures of PSAT1 to identify potential binding pockets

    • Perform in silico screening of compound libraries against the active site

    • Design competitive inhibitors that mimic the transition state of the enzymatic reaction

  • High-throughput screening:

    • Adapt the NADH-coupled spectrophotometric assay for high-throughput format

    • Screen diverse chemical libraries for compounds that inhibit PSAT1 activity

    • Perform secondary assays to confirm specificity and mechanism of action

  • Allosteric modulator development:

    • Identify allosteric sites that regulate PSAT1 activity

    • Design compounds that stabilize inactive conformations

    • Exploit potential differences between canonical and non-canonical functions

  • Targeting PSAT1 stability:

    • Investigate novel stabilization mechanisms, particularly relevant for brain tumors

    • Develop compounds that promote PSAT1 degradation

    • Utilize proteolysis-targeting chimeras (PROTACs) approach

  • Pathway-based targeting:

    • Develop combination approaches targeting multiple enzymes in the serine synthesis pathway

    • Investigate synergistic effects with inhibitors of related metabolic pathways

    • Consider downstream consequences on α-ketoglutarate levels and associated signaling

These approaches align with PSAT1's proposed role as a promising target for tumor suppression in colorectal, esophageal, and breast cancers , as well as potential applications in treating tuberculosis through inhibition of the serine production pathway .

How can researchers effectively measure serine and related metabolites in clinical samples from PSAT1-deficient patients?

Effective measurement of serine and related metabolites in clinical samples requires standardized methodologies:

  • Sample collection and processing:

    • Collect blood samples in appropriate anticoagulants (EDTA or heparin)

    • Process cerebrospinal fluid samples immediately to prevent degradation

    • Consider flash-freezing tissue samples for metabolomic analysis

  • Amino acid analysis:

    • Use high-performance liquid chromatography (HPLC) for separation

    • Couple with mass spectrometry for accurate identification and quantification

    • Include internal standards for normalization and quality control

  • Comprehensive metabolomic profiling:

    • Employ untargeted metabolomics to identify unexpected metabolic alterations

    • Focus on serine synthesis pathway intermediates and related compounds

    • Monitor glycine levels, which are often decreased in conjunction with serine

  • Temporal considerations:

    • Collect samples at consistent times relative to meals to minimize variability

    • Consider longitudinal sampling to track disease progression or treatment response

    • Record fasting status and recent medication use that might affect results

  • Comparative analysis:

    • Establish age and sex-matched control reference ranges

    • Compare results with established thresholds for serine deficiency

    • Correlate metabolite levels with clinical severity and genotype

This methodological approach has been used successfully to characterize biochemical abnormalities in PSAT1-deficient patients, with findings of low plasma and cerebrospinal fluid concentrations of serine and glycine serving as key diagnostic markers .

What methodological considerations are important when studying potential therapeutic approaches for PSAT1-related disorders?

When studying therapeutic approaches for PSAT1-related disorders, researchers should consider:

  • Supplementation strategies assessment:

    • Design protocols for L-serine supplementation with standardized dosing

    • Monitor both biochemical (serine levels) and clinical outcomes

    • Establish appropriate timeframes for evaluating acute versus long-term effects

  • Enzymatic replacement therapy development:

    • Engineer recombinant PSAT1 with appropriate modifications for stability and tissue targeting

    • Develop delivery systems that can cross the blood-brain barrier

    • Establish preclinical models to test efficacy and safety

  • Gene therapy approaches:

    • Design viral vectors for efficient PSAT1 gene delivery

    • Evaluate tissue-specific promoters for targeted expression

    • Assess integration-free approaches to minimize insertional mutagenesis risk

  • Precision medicine considerations:

    • Stratify patients based on mutation type and residual enzyme activity

    • Design mutation-specific therapies (e.g., chaperones for folding mutants)

    • Consider individualized treatment protocols based on biochemical profiles

  • Clinical trial design:

    • Develop appropriate outcome measures sensitive to disease progression

    • Incorporate both objective (biochemical, electrophysiological) and subjective (quality of life) endpoints

    • Consider innovative trial designs to accommodate rare disease populations

These methodological considerations are essential for developing effective treatments for the spectrum of PSAT1-related disorders, from severe Neu-Laxova syndrome to milder juvenile-onset neuropathy, potentially translating recent advances in understanding PSAT1 biology into clinical benefits.

Product Science Overview

Function and Mechanism

PSAT1 catalyzes the reversible conversion of 3-phosphohydroxypyruvate to phosphoserine, which is the second step in the de novo serine synthesis pathway . This reaction is essential for the production of serine, a non-essential amino acid that serves as a precursor for several biomolecules, including glycine, cysteine, and sphingolipids .

The enzyme requires pyridoxal-5’-phosphate (PLP) as a cofactor to facilitate the transfer of amino groups . The activity of PSAT1 is measured by its ability to produce 3-phosphooxypyruvate, with a specific activity greater than 130 pmol/min/μg under specified conditions .

Structure and Expression

Recombinant human PSAT1 is typically expressed in Escherichia coli (E. coli) and is purified with a C-terminal 6-His tag for ease of purification and detection . The recombinant protein has a predicted molecular mass of approximately 41 kDa, although it may appear as a 40 kDa band on SDS-PAGE under reducing conditions .

Clinical Significance

Mutations in the PSAT1 gene can lead to phosphoserine aminotransferase deficiency, a rare metabolic disorder characterized by low levels of serine and glycine in the blood and cerebrospinal fluid . This condition can result in severe neurological symptoms, including seizures, microcephaly, and psychomotor retardation .

Additionally, altered expression of PSAT1 has been implicated in various cancers. Overexpression of PSAT1 has been observed in certain types of cancer, suggesting that the enzyme may play a role in tumor growth and survival .

Applications in Research

Recombinant human PSAT1 is widely used in biochemical and medical research to study the serine biosynthesis pathway and its regulation . It is also utilized in the development of therapeutic strategies for conditions associated with serine deficiency and in cancer research to explore potential targets for drug development .

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.