GMPS Human

GMPS Human Recombinant
Shipped with Ice Packs
In Stock

Description

Enzymatic Function and Reaction Mechanism

GMPS operates through two distinct catalytic domains:

  • Glutaminase (GATase) domain: Hydrolyzes glutamine to generate ammonia .

  • Synthetase (ATPPase) domain: Binds ATP and XMP, forming an adenyl-XMP intermediate before ammonia incorporation yields GMP .

The reaction is summarized as:

ATP + XMP + L-glutamineAMP + diphosphate + GMP + L-glutamate\text{ATP + XMP + L-glutamine} \rightarrow \text{AMP + diphosphate + GMP + L-glutamate}

In vitro, GMPS can also utilize exogenous ammonia, but physiological activity relies on intramolecular ammonia channeling from the GATase domain .

Domain Architecture

  • N-terminal GATase domain (residues 1–206): Contains a catalytic triad (Cys86, His181, Glu183) for glutamine hydrolysis .

  • C-terminal ATPPase domain: Includes a P-loop (residues 235–241) for ATP binding and specificity .

Quaternary Structure

Human GMPS forms a dimer in crystal structures, with subunit interactions mediated by a 130-residue insert absent in bacterial homologs. This insert facilitates substrate binding and dimer stabilization .

Purine Biosynthesis

GMPS is the second enzyme in the IMP-to-GMP pathway:

  1. IMP dehydrogenase oxidizes IMP to XMP.

  2. GMPS aminates XMP to GMP, which is further phosphorylated to GTP .

Clinical Relevance

  • Drug target: GMPS is explored for inhibitors in proliferative diseases. Psicofuranin, a nucleoside analog, inhibits GMPS activity .

  • Cancer therapy: Elevated GMPS activity in tumors necessitates nucleotide analogs to disrupt DNA synthesis .

Comparative Analysis Across Species

Human GMPS diverges from bacterial homologs in oligomerization and regulation:

  • Allosteric regulation: ATP and XMP binding activate the GATase domain, ensuring coordinated catalysis .

  • Structural dynamics: The LID motif in human GMPS undergoes conformational changes to sequester XMP during adenylation .

Product Specs

Introduction
GMP synthase (GMPS) is an essential enzyme in the purine biosynthesis pathway, responsible for catalyzing the final step in GMP production. This enzyme, a homodimer, converts XMP to GMP through an ATP-dependent amination reaction. GMPS consists of two domains: a GMP-binding domain and a glutamine amidotransferase type-1 domain, the latter being crucial for its catalytic function. This enzyme plays a critical role in de novo nucleotide synthesis, providing the building blocks for DNA and RNA synthesis. Additionally, GMPS contributes to the production of GTP, a molecule involved in various cellular processes essential for cell division. Notably, chromosomal translocations involving the GMPS gene have been observed in acute myeloid leukemia cases, suggesting a potential link between GMPS and cancer development.
Description
Recombinant human GMPS, expressed in E. coli, is a non-glycosylated polypeptide chain with 717 amino acids (including a 24 amino acid His-tag at the N-terminus). This protein, with a molecular weight of 79.2 kDa, is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution, sterile-filtered.
Formulation
The GMPS solution is provided at a concentration of 0.5 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 1 mM DTT, 30% glycerol, and 0.1 M NaCl.
Stability
For short-term storage (2-4 weeks), the GMPS solution should be stored at 4°C. For extended storage, it is recommended to store the solution at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. Avoid repeated freeze-thaw cycles to maintain protein stability.
Purity
The purity of GMPS is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms

GMP synthase [glutamine-hydrolyzing], GMP synthetase, Glutamine amidotransferase, GMPS, GMP synthase, guanosine 5'-monophosphate synthase, MLL/GMPS fusion protein.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMALCNG DSKLENAGGD LKDGHHHYEG AVVILDAGAQ YGKVIDRRVR ELFVQSEIFP LETPAFAIKE QGFRAIIISG GPNSVYAEDA PWFDPAIFTI GKPVLGICYG MQMMNKVFGG TVHKKSVRED GVFNISVDNT CSLFRGLQKE EVVLLTHGDS
VDKVADGFKV VARSGNIVAG IANESKKLYG AQFHPEVGLT ENGKVILKNF LYDIAGCSGT FTVQNRELEC IREIKERVGT SKVLVLLSGG VDSTVCTALL NRALNQEQVI AVHIDNGFMR KRESQSVEEA LKKLGIQVKV INAAHSFYNG TTTLPISDED RTPRKRISKT LNMTTSPEEK
RKIIGDTFVK IANEVIGEMN LKPEEVFLAQ GTLRPDLIES ASLVASGKAE LIKTHHNDTE LIRKLREEGK VIEPLKDFHK DEVRILGREL GLPEELVSRH PFPGPGLAIR VICAEEPYIC KDFPETNNIL KIVADFSASV KKPHTLLQRV KACTTEEDQE KLMQITSLHS LNAFLLPIKT
VGVQGDCRSY SYVCGISSKD EPDWESLIFL ARLIPRMCHN VNRVVYIFGP PVKEPPTDVT PTFLTTGVLS TLRQADFEAH NILRESGYAG KISQMPVILT PLHFDRDPLQ KQPSCQRSVV IRTFITSDFM TGIPATPGNE IPVEVVLKMV TEIKKIPGIS RIMYDLTSKP PGTTEWE.

Q&A

What are the fundamental requirements for establishing a GMP-compliant human stem cell laboratory?

Establishing a GMP-compliant stem cell laboratory requires meeting strict requirements defined by regulatory bodies like the FDA and adherence to CFR Title 21. The laboratory design must carefully account for product flow and human interactions to ensure safety, identity, purity, and potency of manufactured products . Specifically, the laboratory layout should enable various specialized processes including:

  • Generation of clinical grade peptide-based products

  • Production of gene-modified cellular products

  • Cryopreservation of manufactured products

  • Quality control testing facilities

The design must also consider employee well-being, creating an environment where investigators can work comfortably and efficiently without fatigue. This includes appropriate lighting, ergonomic workstations, and proper ventilation systems . Additionally, the design should facilitate inspection and monitoring processes required for GMP compliance while maintaining workflow efficiency.

How should researchers structure GMP training programs for personnel working with human biological materials?

Regulatory agencies worldwide require that GMP training be conducted regularly for all personnel involved in producing pharmaceutical and biological products, with documented evidence of both completion and effectiveness . An effective GMP training program should include:

  • Written programs specific to different personnel categories (production, laboratory, maintenance, cleaning)

  • Initial orientation training for newly recruited personnel

  • Ongoing training with periodic assessment of practical effectiveness

  • Specialized training for personnel working in contamination-sensitive areas

Training sessions should comprehensively discuss quality assurance concepts and implementation measures. All training programs must be approved by appropriate department heads (production or quality control) with detailed records maintained . According to regulatory guidelines, even outside contractors or maintenance staff who enter production areas must receive appropriate instruction and supervision to maintain GMP compliance.

How can researchers validate alternative methods for quality control in GMP-compliant human cell manufacturing?

The European Pharmacopoeia (since July 2007) permits replacing official methods with Nucleic Acid Amplification techniques after suitable validation . A methodological approach to validation includes:

  • Compliance with international GMP requirements and standards (e.g., European Pharmacopoeia sec. 2.6.7 and International Conference on Harmonization Q2)

  • Systematic evaluation of:

    • Sensitivity (lower detection limits)

    • Matrix-related interferences

    • Specificity (ability to distinguish target from similar organisms)

    • Robustness (reliability under varying conditions)

For example, in mycoplasma detection for mesenchymal stromal cells, validation of real-time PCR methods should include interference removal strategies as matrix composition can affect PCR reactions . Complete validation documentation should demonstrate that the alternative method is at least equivalent to the official method in terms of detection capability.

What methodological approaches should be implemented for investigating and documenting deviations in GMP-compliant human cell therapy processes?

Effective deviation management in GMP environments requires a systematic approach to problem investigation and documentation. Methodologically, researchers should:

Organizations that provide extensive training to key personnel on investigating deviations and writing reports show significant reduction in recurring problems . The investigation should involve a multidisciplinary team with representation from quality assurance, production, and relevant technical areas to ensure comprehensive assessment.

What is the role of GMPS in human cellular metabolism and proliferation?

GMPS (Guanosine Monophosphate Synthetase) plays a crucial role in de novo purine biosynthesis by catalyzing the final step in guanine nucleotide synthesis. Specifically, it converts xanthine monophosphate (XMP) to guanosine monophosphate (GMP) through an amination reaction that requires glutamine and ATP . The enzyme has a complex structure consisting of:

  • An N-terminal glutaminase (GATase) domain that binds glutamine and generates ammonia

  • A C-terminal synthetase (ATPPase) domain where the ammonia aminates XMP to generate GMP

This reaction is essential for maintaining the cellular guanine nucleotide pool, which is critical for DNA and RNA synthesis during cell proliferation. The importance of GMPS in cellular metabolism is evidenced by the significant growth inhibition observed in cancer cell lines when GMPS is inhibited pharmacologically or through knockdown approaches .

How does GMPS expression relate to human disease progression?

In experimental models, GMPS has been shown to be overexpressed in metastatic human melanoma cells, where its inhibition suppresses melanoma cell invasion and tumorigenicity . Similar findings in prostate cancer cell lines (LNCaP and PC-3) demonstrate that pharmacological inhibition or knockdown of GMPS significantly decreases cell growth, highlighting its importance in cancer cell proliferation.

These findings collectively suggest that GMPS may serve as both a prognostic marker and potential therapeutic target in various cancers, warranting further investigation into its regulatory mechanisms and clinical applications.

How do alterations in GMPS expression affect purine biosynthesis pathways in human cancer cells?

GMPS knockdown induces complex metabolic adaptations in purine biosynthesis pathways. When GMPS is inhibited, cancer cells demonstrate compensatory changes in metabolic flux through the purinosome complex, which increases activity under low purine conditions . Detailed metabolic analysis using 15N-(amide)-glutamine tracing reveals:

MetaboliteEffect in shGMPS cellsTimepointMechanism
IMPIncreased 15N-amide labeling2 hoursIncreased purinosome activity
GMPDecreased 15N labeling (20% vs 28% in control)2 hoursReduced conversion from XMP due to low GMPS
GuanineDecreased 15N labeling (14% vs 21% in control)2 hoursConsequence of reduced GMP synthesis

This metabolic reprogramming illustrates the dynamic nature of nucleotide biosynthesis pathways and highlights the critical role of GMPS as a regulatory point in guanine nucleotide production.

What methodological approaches should be considered when designing rescue experiments to distinguish between direct and indirect effects of GMPS inhibition?

Designing effective rescue experiments requires careful methodological considerations to differentiate between direct effects on nucleotide synthesis and other potential cellular roles of GMPS. A comprehensive experimental approach should include:

  • Exogenous nucleotide supplementation:

    • Addition of guanosine (typically 200 μM) to activate the salvage pathway

    • Measurement of growth recovery percentages (e.g., 80% in LNCaP; 78% in PC-3 as shown in previous studies)

    • Determination of dose-response relationships for rescue compounds

  • Metabolic tracing experiments:

    • Utilize isotope-labeled precursors (15N-(amide)-glutamine, 13C-glutamine)

    • Time-course analysis (2h, 24h) to capture both immediate and adaptive responses

    • Quantification of labeled vs. unlabeled metabolite ratios

  • Mechanistic validation:

    • Complementary inhibition of salvage pathway enzymes

    • Introduction of GMPS mutants with selective functional deficits

    • Assessment of downstream signaling pathways

Incomplete rescue (typically 75-80%) suggests either insufficiency of salvage pathways to match normal de novo synthesis rates or additional GMPS cellular functions beyond nucleotide synthesis . The systematic application of these approaches enables researchers to delineate the precise mechanisms through which GMPS inhibition affects cellular function, guiding the development of targeted therapeutic strategies.

How can GMP principles be applied to experimental design when studying GMPS inhibition in primary human cells?

Integrating GMP principles into GMPS research with primary human cells requires special considerations for quality, reproducibility, and safety. Researchers should implement:

  • Standardized cell isolation and culture protocols with:

    • Validated reagent quality testing

    • Documented cell characterization procedures

    • Traceability systems for all materials and processes

  • Quality control measures including:

    • Mycoplasma testing using validated PCR-based methods as demonstrated with mesenchymal stromal cells

    • Cell identity verification through appropriate markers

    • Functional assays to confirm typical cell behaviors

  • Process validation approaches:

    • Determination of critical process parameters affecting GMPS expression or activity

    • Establishment of acceptance criteria for experimental outcomes

    • Implementation of environmental monitoring where appropriate

This methodological integration ensures that research findings on GMPS inhibition in primary human cells are reliable, reproducible, and potentially translatable to clinical applications. The implementation of GMP principles early in research helps facilitate smoother transitions to later-stage clinical development of any therapeutic strategies targeting GMPS.

What are the key methodological considerations for developing GMPS-targeted therapies in GMP-compliant manufacturing settings?

Developing GMPS-targeted therapies requires bridging basic research findings with GMP manufacturing requirements. Key methodological considerations include:

  • Target validation and specificity:

    • Comprehensive profiling of GMPS expression across normal and disease tissues

    • Evaluation of off-target effects through proteomics and metabolomics

    • Establishment of therapeutic windows based on differential expression

  • Manufacturing process development:

    • Design of laboratory spaces accounting for product flow and human interactions

    • Implementation of risk-based quality management systems

    • Development of analytical methods for product characterization

  • Translational considerations:

    • Correlation of preclinical findings with clinical parameters

    • Development of companion diagnostics to identify appropriate patient populations

    • Establishment of release criteria based on critical quality attributes

The integration of these methodological approaches ensures that GMPS-targeted therapies can be manufactured consistently and safely while maintaining their therapeutic efficacy. Early consideration of GMP requirements during development can significantly accelerate the translation of research findings into clinical applications.

Product Science Overview

Introduction

Guanosine monophosphate synthetase (GMPS) is an enzyme that plays a crucial role in the de novo synthesis of guanosine monophosphate (GMP) from xanthosine monophosphate (XMP). This enzyme is part of the glutamine amidotransferase (GAT) family and is essential for various cellular processes, including DNA replication, transcription, and translation .

Biological Properties

GMPS is a modular enzyme with distinct domains responsible for its catalytic activities. It catalyzes the hydrolysis of glutamine and transfers the generated ammonia to XMP, facilitating the synthesis of GMP. The enzyme’s structure includes an ammonia channel that connects the two catalytic sites, allowing efficient transfer of ammonia .

Expression Patterns and Tissue Distribution

GMPS is expressed in various tissues, with significant expression in the liver, kidney, and brain. The Human Protein Atlas provides a detailed overview of GMPS expression across different tissues, highlighting its intracellular localization, primarily in the cytosol .

Biological Functions

GMPS is vital for the synthesis of GMP, which is a precursor to guanosine triphosphate (GTP). GTP is essential for numerous cellular functions, including signal transduction, protein synthesis, and cell division. The enzyme’s activity is regulated by allosteric mechanisms, ensuring precise control of GMP synthesis .

Modes of Action

The catalytic mechanism of GMPS involves two distinct activities: glutaminolysis and amination. The enzyme’s structure facilitates the coordination of these activities through conformational changes and the ammonia channel. This efficient mechanism ensures the continuous supply of GMP for cellular processes .

Regulatory Mechanisms

The expression and activity of GMPS are tightly regulated to maintain cellular homeostasis. Various factors, including feedback inhibition by GMP and GTP, modulate the enzyme’s activity. Additionally, post-translational modifications and interactions with other proteins contribute to the regulation of GMPS .

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.