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:
In vitro, GMPS can also utilize exogenous ammonia, but physiological activity relies on intramolecular ammonia channeling from the GATase domain .
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 .
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 .
GMPS is the second enzyme in the IMP-to-GMP pathway:
IMP dehydrogenase oxidizes IMP to XMP.
GMPS aminates XMP to GMP, which is further phosphorylated to GTP .
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 .
Human GMPS diverges from bacterial homologs in oligomerization and regulation:
GMP synthase [glutamine-hydrolyzing], GMP synthetase, Glutamine amidotransferase, GMPS, GMP synthase, guanosine 5'-monophosphate synthase, MLL/GMPS fusion protein.
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.
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.
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.
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.
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 .
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.
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:
Metabolite | Effect in shGMPS cells | Timepoint | Mechanism |
---|---|---|---|
IMP | Increased 15N-amide labeling | 2 hours | Increased purinosome activity |
GMP | Decreased 15N labeling (20% vs 28% in control) | 2 hours | Reduced conversion from XMP due to low GMPS |
Guanine | Decreased 15N labeling (14% vs 21% in control) | 2 hours | Consequence 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.
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:
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.
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:
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.
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:
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.
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 .
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 .
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 .
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 .
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 .