Human Phosphopantothenoylcysteine Synthetase (PPCS) is a critical enzyme in the coenzyme A (CoA) biosynthesis pathway. It catalyzes the second step in this essential pathway, specifically the formation of phosphopantothenoylcysteine from (R)-phosphopantothenate (PPA) and L-cysteine with the concomitant consumption of a nucleotide triphosphate. This reaction is fundamental to cellular metabolism as CoA serves as an essential cofactor for numerous metabolic reactions, including the citric acid cycle, fatty acid synthesis and oxidation, and amino acid metabolism. PPCS is encoded by the human coaB gene and functions as part of the multiprotein CoA synthase complex. Defects in PPCS function have been linked to severe metabolic disorders, including dilated cardiomyopathy, highlighting its central importance in human cellular metabolism .
The Predictability, Computability, and Stability (PCS) framework represents an important methodological approach in data science that has applications across human research domains. This framework embeds scientific principles of prediction and replication in data-driven decision making while recognizing computation's central role. The PCS workflow uses predictability as a reality check for research outcomes and considers computational aspects in data collection, storage, and algorithm design. It further augments these principles with stability assessments to evaluate how human judgment impacts data results through data and model/algorithm perturbations. PCS inference procedures, including perturbation intervals and hypothesis testing, investigate result stability relative to problem formulation, data cleaning, modeling decisions, and interpretations. This framework is particularly valuable for responsible, reliable, reproducible, and transparent analysis across fields of science, social science, engineering, and medical research involving human subjects .
Human PPCS follows a Bi Uni Uni Bi Ping Pong mechanism similar to that previously described for E. faecalis PPCS. Experimentally validating this mechanism requires sophisticated enzyme kinetics approaches:
Reaction Order Analysis: Steady-state kinetic studies using varying concentrations of all three substrates (nucleotide, PPA, and cysteine) help establish the binding order and release of products.
Oxygen Transfer Studies: Experiments using [carboxyl-18O] phosphopantothenate demonstrate that the 18O from phosphopantothenate is incorporated into the AMP or CMP produced during catalysis. This confirms the formation of phosphopantothenoyl cytidylate or phosphopantothenoyl adenylate intermediates, supporting similar catalytic mechanisms under both CTP and ATP conditions.
Cooperativity Assessment: Hill plot analysis reveals a Hill constant of 1.7 for ATP binding, indicating cooperative binding behavior that is not observed with CTP.
The kinetic parameters determined through these methods include:
Substrate | Condition | Km (μM) | kcat (s-1) | Notes |
---|---|---|---|---|
CTP | CTP-based catalysis | 265 | 0.53±0.01 | Non-cooperative binding |
PPA | CTP-based catalysis | 57 | 0.53±0.01 | - |
Cysteine | CTP-based catalysis | 16 | 0.53±0.01 | - |
ATP | ATP-based catalysis | 269 | 0.56 | Cooperative binding (Hill constant = 1.7) |
PPA | ATP-based catalysis | 13 | 0.56 | - |
Cysteine | ATP-based catalysis | 14 | 0.56 | - |
These detailed kinetic studies provide mechanistic insights into how human PPCS functions and can guide the development of inhibitors or activators with potential therapeutic applications .
The crystal structure of human PPCS in complex with phosphorylated hopantenate (P-HoPan) and AMPPNP (a non-hydrolyzable ATP analog) provides critical insights into the enzyme's catalytic mechanism and substrate binding modes. This structure (PDB ID: 7EDZ) has several important research implications:
Substrate Binding Site Elucidation: The structure reveals precisely how the phosphopantothenate substrate (or its analog P-HoPan) binds within the active site, identifying key residues involved in substrate recognition and positioning.
Nucleotide Binding Mode: The co-crystallized AMPPNP demonstrates the binding mode of the activating nucleotide, clarifying how ATP interacts with the enzyme during catalysis.
Cysteine Binding Site Identification: Perhaps most significantly, this structure helps elucidate the previously unknown binding mode for cysteine, the third substrate in the reaction. This represents a major advance in understanding the complete catalytic mechanism.
Disease-Relevant Mutations: The structure provides molecular context for understanding how mutations in human PPCS linked to severe dilated cardiomyopathy affect enzyme function, by revealing their structural positions relative to catalytic and substrate-binding sites.
Inhibitor Design Platform: The structure serves as a crucial template for structure-based drug design efforts targeting PPCS, potentially leading to novel therapeutics for diseases involving CoA metabolism disruption.
This structural data, combined with functional analyses, demonstrates that P-HoPan forms a nonproductive substrate complex with PPCS, explaining its inhibitory mechanism and providing a framework for understanding the enzyme's catalytic cycle at atomic resolution .
Investigating PPCS in Pantothenate Kinase-Associated Neurodegeneration (PKAN) and related neurological disorders requires a multi-faceted methodological approach:
Enzyme Kinetic Studies: Detailed characterization of PPCS activity using purified enzymes (wild-type and disease-associated variants) helps establish mechanistic understanding. The human coaB gene encoding PPCS can be cloned into expression vectors (e.g., pET23a) and overexpressed in E. coli BL21(DE3) to yield approximately 10mg of purified enzyme per liter of culture, enabling comprehensive biochemical analysis .
Crystal Structure Analysis: Structural studies like the 7EDZ PPCS-P-HoPan-AMPPNP complex provide critical insights into how inhibitors and disease-associated mutations affect enzyme function. These structural analyses reveal binding modes and potential mechanisms of enzyme dysfunction .
Cell Biology Models: PKAN cell models treated with coenzyme A pathway modulators like hopantenate (HoPan) help elucidate disease mechanisms. Recent research has shown that contrary to previous assumptions, HoPan does not directly inhibit pantothenate kinase (PanK) but rather is phosphorylated by PanK and subsequently inhibits PPCS through formation of a nonproductive substrate complex .
Metabolic Profiling: Comprehensive analysis of CoA-related metabolites in patient samples or model systems using liquid chromatography-mass spectrometry helps identify biomarkers and understand metabolic consequences of PPCS dysfunction.
Genetic Analysis: Sequencing the PPCS gene in patient cohorts with unexplained neurological symptoms similar to PKAN can identify novel disease-associated variants.
Imaging Studies: Advanced neuroimaging techniques in combination with biochemical analyses provide correlations between PPCS dysfunction, CoA levels, and progressive neurodegeneration.
These complementary approaches provide a comprehensive understanding of PPCS's role in neurological disorders, potentially leading to novel therapeutic strategies targeting specific steps in the CoA biosynthesis pathway .
Oxygen transfer studies represent a powerful approach for investigating the mechanistic details of human PPCS catalysis. To optimize these studies for maximum insight:
Isotope Labeling Strategy: Use [carboxyl-18O] phosphopantothenate as the primary labeled substrate. The strategic placement of the 18O label at the carboxyl group is critical as this oxygen is involved in the formation of the phosphopantothenoyl adenylate or cytidylate intermediate.
Reaction Condition Optimization: Carefully control reaction conditions including pH (typically 7.0-7.5), temperature (30-37°C), and buffer composition to ensure optimal enzyme activity while maintaining stability of the isotope-labeled substrate.
Product Isolation Techniques: Develop efficient methods for isolating the nucleotide monophosphate products (AMP or CMP) from reaction mixtures, typically using HPLC with appropriate columns for nucleotide separation.
Mass Spectrometric Analysis: Employ high-resolution mass spectrometry (preferably with techniques like isotope ratio MS) to quantitatively analyze the incorporation of 18O into the AMP or CMP products. This requires careful calibration using unlabeled standards.
Parallel Reaction Monitoring: Conduct parallel reactions using ATP and CTP as the nucleotide substrate to compare oxygen transfer patterns between the two catalytic modes.
Control Reactions: Include appropriate controls such as reactions with unlabeled substrates and heat-inactivated enzyme to account for any non-enzymatic oxygen exchange.
Data Analysis Framework: Develop a quantitative framework for analyzing isotope incorporation data that can distinguish between different mechanistic possibilities.
These studies have successfully demonstrated that 18O from phosphopantothenate is incorporated into the AMP or CMP produced during PPCS catalysis, providing strong evidence for the formation of phosphopantothenoyl cytidylate or phosphopantothenoyl adenylate intermediates. This confirms similar catalytic mechanisms under both CTP and ATP conditions despite differences in cooperativity .
Resolving contradictory findings regarding HoPan's inhibitory mechanism in the CoA biosynthesis pathway requires a comprehensive, multi-method approach:
Sequential Enzyme Analysis: Individually assess HoPan's effects on each enzyme in the CoA biosynthesis pathway (PanK, PPCS, PPCDC, PPAT, and DPCK) using purified enzymes and appropriate activity assays. This isolates each step to determine where inhibition occurs.
Metabolite Profiling: Apply metabolomics techniques to identify accumulating intermediates in HoPan-treated cells or tissues. The detection of phosphorylated HoPan and reduced levels of phosphopantothenoylcysteine would support PPCS as the primary target.
Structural Biology Approaches: Obtain crystal structures of PanK and PPCS in complex with HoPan or P-HoPan (as accomplished with the 7EDZ structure). Direct visualization of binding modes provides definitive evidence of interaction sites.
Kinetic Mechanism Determination: Perform detailed enzyme kinetic studies comparing substrate competition patterns between natural substrates and HoPan. Determine if HoPan acts as a competitive, noncompetitive, or uncompetitive inhibitor.
Genetic Validation: Use CRISPR-Cas9 to create cell lines with altered expression of PanK or PPCS to determine if sensitivity to HoPan correlates with expression levels of either enzyme.
In Cellulo Metabolic Bypass Experiments: Attempt to rescue HoPan inhibition by supplementing cells with downstream metabolites (like phosphopantothenoylcysteine). Rescue would confirm the inhibition site.
Domain-Specific Mutagenesis: Create targeted mutations in PPCS and PanK at putative HoPan binding sites to identify residues critical for inhibitor interaction.
Through such comprehensive approaches, researchers have resolved earlier contradictions by demonstrating that HoPan is actually phosphorylated by PanK (rather than inhibiting it) and subsequently inhibits PPCS through formation of a nonproductive substrate complex. This finding significantly revised the understanding of HoPan's mechanism of action in modulating CoA levels, particularly important for PKAN disease models .
When investigating Posterior Parietal Cortex (PPC) specificity for human reach planning, several critical experimental design considerations must be addressed:
Precise Anatomical Targeting: The human PPC contains multiple functional subregions including superior parieto-occipital cortex (SPOC), angular gyrus (AG), and midposterior intraparietal sulcus (mIPS). Experimental designs must precisely target these specific areas using anatomical landmarks and functional localization.
Causal Interference Methodology: Transcranial Magnetic Stimulation (TMS) provides a powerful approach for establishing causal relationships between brain regions and functions. Online repetitive TMS (rTMS) at 10 Hz has been successfully employed to examine effector specificity in PPC subregions.
Task Design Considerations:
Include both reach and saccade conditions to test effector specificity
Use delayed response paradigms to separate planning from execution phases
Present targets across multiple spatial locations (e.g., horizontally aligned targets at varying visual angles)
Control for visual feedback conditions by testing in both darkness and with visual feedback
Include both hands to examine limb specificity
Performance Metrics:
Measure both accuracy (constant errors: signed difference between mean endpoint and target positions)
Assess precision (variable errors: area of 95% confidence ellipses fitted to endpoint distributions)
Control Conditions:
Include sham stimulation
Test multiple PPC sites to establish regional specificity
Control for non-specific TMS effects
Statistical Approach: Implement appropriate statistical designs to test for interactions between stimulation site, effector type (reach vs. saccade), visual feedback condition, and target location.
Research using these approaches has established a caudorostral gradient of effector preferences in human PPC, with SPOC showing specificity for reach planning only, while anterior-lateral parietal areas mIPS and AG are involved in encoding both saccade and reach movements. These findings extend our understanding of the neuroscience of human visuomotor transformations and highlight the importance of rigorous experimental design in establishing functional specificity in human brain regions .
Current research on human PPCS faces several important limitations that future studies should address:
Limited Structural Data on Reaction Intermediates: While the 7EDZ structure provides insights into substrate binding, structures capturing different catalytic states (particularly the phosphopantothenoyl-adenylate or cytidylate intermediates) are lacking. Future studies should aim to trap and crystallize these intermediates using substrate analogs or catalytically inactive mutants.
Incomplete Understanding of Regulatory Mechanisms: How PPCS activity is regulated in different physiological and pathological contexts remains poorly characterized. Future studies should investigate potential post-translational modifications, protein-protein interactions, and allosteric regulation mechanisms that may modulate PPCS function.
Limited In Vivo Data: Most PPCS studies rely on in vitro biochemical approaches with purified enzymes. More sophisticated in vivo models, including humanized mouse models or human cell lines with engineered PPCS variants, would provide physiologically relevant insights into PPCS function and dysfunction.
Therapeutic Development Challenges: Despite knowledge of PPCS's role in CoA metabolism and implications in diseases like cardiomyopathy, translating this understanding into therapeutic approaches remains challenging. Structure-guided drug design efforts should be intensified to develop specific PPCS modulators.
Technical Limitations in Activity Assays: Current PPCS activity assays have limitations in sensitivity and throughput. Development of more sensitive, high-throughput assays would facilitate larger-scale screening efforts and more detailed kinetic analyses.
Tissue-Specific Expression and Function: The tissue-specific roles of PPCS and potential isoform-specific functions remain underexplored. Future studies should investigate tissue-specific expression patterns and potential specialized roles in tissues with high metabolic demands.
Integration with Systems Biology: Current understanding of PPCS tends to focus on the enzyme in isolation rather than its role within broader metabolic networks. Systems biology approaches integrating transcriptomics, proteomics, and metabolomics would provide a more comprehensive understanding of PPCS in cellular metabolism.
Addressing these limitations will require interdisciplinary approaches combining structural biology, enzymology, cell biology, genetics, and systems biology to develop a more complete understanding of human PPCS in health and disease .
The Predictability, Computability, and Stability (PCS) framework offers significant potential for enhancing reproducibility in PPCS enzyme research:
Predictability for Experimental Validation: The predictability principle can guide experimental design by establishing clear prediction tasks for PPCS research. For example, predicting how specific mutations will affect enzyme kinetics allows for direct experimental validation, creating a feedback loop that strengthens research reliability.
Computational Modeling Standards: PCS emphasizes computability, which can standardize approaches to PPCS structural and functional modeling. Implementing consistent computational protocols for tasks such as molecular dynamics simulations of PPCS-substrate interactions would improve cross-study comparability.
Stability Analysis for Experimental Conditions: The stability principle is particularly valuable for biochemical research, where small variations in experimental conditions can significantly impact results. By systematically perturbing experimental parameters (pH, temperature, buffer composition) and analyzing the stability of PPCS activity measurements, researchers can identify robust experimental protocols.
Documentation and Workflow Standards: PCS workflow documentation (using R Markdown or Jupyter Notebook) provides a framework for transparent reporting of data processing and analysis in PPCS research. This ensures that data transformations, statistical methods, and visualization techniques are fully documented and reproducible.
PCS Inference for Reliability Assessment: PCS inference procedures, including perturbation intervals and hypothesis testing, can be applied to assess the stability of PPCS research findings relative to:
Sample preparation variations
Enzyme purification methods
Data cleaning decisions
Kinetic model selection
Statistical analysis choices
Meta-Analysis Framework: The PCS approach provides a structured framework for comparing results across different PPCS studies, helping to resolve contradictions and identify sources of variability in the literature.
Preregistration Templates: Development of PCS-based preregistration templates specifically for PPCS enzymatic studies would reduce researcher degrees of freedom and publication bias.
Implementing the PCS framework in PPCS research would create more responsible, reliable, reproducible, and transparent results across the research lifecycle, from hypothesis generation to experimental design, data analysis, and interpretation .
Research on human PPCS has significant implications for understanding various metabolic disorders beyond Pantothenate Kinase-Associated Neurodegeneration (PKAN):
Dilated Cardiomyopathy: Recent structural analyses of human PPCS have revealed the functional implications of mutations linked to severe dilated cardiomyopathy. The 7EDZ crystal structure provides mechanistic insights into how these mutations disrupt enzyme function, potentially through altered substrate binding or catalytic efficiency. This connection establishes PPCS as a critical link between CoA metabolism and cardiac function .
Metabolic Syndrome Components: CoA and its derivatives are central to energy metabolism, including fatty acid oxidation and synthesis. PPCS dysfunction could contribute to dyslipidemia, insulin resistance, and other metabolic syndrome components through disrupted CoA-dependent processes. Understanding PPCS regulation may provide novel targets for metabolic disorder treatments.
Neurodegenerative Disorders: While PKAN directly involves pantothenate kinase, the broader role of CoA metabolism in neuronal function suggests PPCS may be relevant to other neurodegenerative conditions. Neurons have high energy demands and rely on proper CoA metabolism for mitochondrial function and neurotransmitter synthesis.
Cancer Metabolism: Rapidly proliferating cancer cells often exhibit altered metabolism, including changes in fatty acid synthesis and mitochondrial function. As a key enzyme in CoA biosynthesis, PPCS may represent an unexplored metabolic vulnerability in certain cancers with high CoA utilization.
Aging-Related Metabolic Decline: Age-related declines in mitochondrial function and metabolic efficiency may involve changes in CoA metabolism. PPCS function across the lifespan could provide insights into metabolic aspects of aging.
Drug-Induced Metabolic Disruptions: Understanding PPCS function and inhibition mechanisms provides a framework for evaluating how certain drugs might inadvertently affect CoA metabolism, potentially explaining some adverse metabolic effects.
Mitochondrial Disorders: Given CoA's central role in mitochondrial metabolism, PPCS research contributes to our understanding of primary and secondary mitochondrial disorders, potentially revealing new therapeutic approaches for these often devastating conditions.
By elucidating the structural, kinetic, and regulatory properties of human PPCS, researchers gain critical insights into CoA metabolism that extend far beyond PKAN, potentially impacting our understanding and treatment of numerous metabolic disorders .
Effectively correlating PPCS structural studies with clinical manifestations requires a translational research approach bridging molecular mechanisms and clinical observations:
Structure-Function Correlation for Disease Mutations:
Map known disease-associated mutations onto the high-resolution PPCS crystal structure (like 7EDZ)
Perform in silico analyses to predict effects on protein stability, catalytic efficiency, and substrate binding
Validate predictions through site-directed mutagenesis and enzymatic assays
Create a comprehensive database linking structural positions, biochemical effects, and clinical phenotypes
Development of Functional Biomarkers:
Design assays measuring specific metabolic intermediates affected by PPCS dysfunction
Correlate metabolite levels with disease severity and progression
Validate biomarkers in patient cohorts with different PPCS variants
Patient-Derived Models:
Generate induced pluripotent stem cells (iPSCs) from patients with PPCS mutations
Differentiate into relevant cell types (cardiomyocytes, neurons) for functional studies
Compare cellular phenotypes with biochemical and structural predictions
Test targeted interventions based on structural insights
Tissue-Specific Considerations:
Investigate why certain PPCS mutations predominantly affect specific tissues (e.g., heart in dilated cardiomyopathy)
Examine tissue-specific expression patterns and potential interaction partners
Analyze tissue-specific metabolic demands for CoA and its derivatives
Genotype-Phenotype Correlation Studies:
Conduct comprehensive clinical characterization of patients with PPCS variants
Correlate specific mutations with disease onset, progression, and response to interventions
Use structural data to classify mutations functionally (e.g., catalytic site vs. substrate binding vs. protein stability)
Therapeutic Development Pipeline:
Use structural insights to design targeted interventions for specific mutation types
Develop high-throughput screening assays based on structural knowledge
Test candidate compounds in patient-derived cellular models
Design mutation-specific therapeutic approaches
Multi-Omics Integration:
Combine structural insights with proteomics, metabolomics, and transcriptomics data
Create integrated models explaining how PPCS structural alterations lead to downstream metabolic and clinical effects
Identify potential compensatory mechanisms in different genetic backgrounds
This comprehensive approach allows researchers to establish mechanistic links between PPCS structural features and clinical manifestations, ultimately leading to more precise diagnosis, prognosis, and treatment strategies for patients with PPCS-related disorders .
The field of human PPCS research is poised for significant advances in the coming five years, with several promising research directions:
Structural Biology Breakthroughs: Building on the 7EDZ crystal structure, researchers will likely pursue structures of human PPCS in different conformational states, with various substrates/products, and disease-causing mutations. Cryo-EM approaches may reveal dynamic aspects of PPCS function not captured in crystal structures, particularly regarding the cooperative binding mechanism observed with ATP.
Systems Biology Integration: More comprehensive integration of PPCS into broader metabolic networks will emerge, with particular focus on tissue-specific CoA metabolism regulation. Multi-omics approaches will elucidate how PPCS activity influences and responds to changing metabolic states in different physiological and pathological contexts.
Precision Medicine Applications: Advances in understanding the molecular consequences of PPCS mutations will enable more personalized approaches to treating PPCS-related disorders. This may include mutation-specific therapies or interventions targeting specific metabolic consequences of PPCS dysfunction.
Novel Regulatory Mechanisms: Discovery of previously uncharacterized regulatory mechanisms controlling PPCS activity, including potential allosteric regulators, post-translational modifications, and protein-protein interactions that modulate enzyme function in response to cellular needs.
Therapeutic Development: Structure-guided drug design efforts will yield more specific and potent modulators of PPCS activity, potentially leading to first-in-class therapeutics for disorders involving CoA metabolism. These may include both activators and inhibitors tailored to specific clinical contexts.
Advanced In Vivo Models: Development of more sophisticated animal models with humanized PPCS or conditional PPCS variants will provide better platforms for understanding tissue-specific roles and testing therapeutic interventions before clinical translation.
Clinical Biomarker Validation: Identification and validation of PPCS-related biomarkers that reflect enzyme activity in vivo will improve diagnosis, monitoring, and treatment assessment for related disorders.
Synthetic Biology Applications: Engineering of PPCS variants with novel properties for biotechnology applications, potentially including production of CoA derivatives or related compounds of industrial or pharmaceutical interest.
These research directions will collectively advance our understanding of human PPCS biology while developing translational applications that address the clinical challenges associated with CoA metabolism disorders .
Interdisciplinary collaboration can significantly enhance human PPCS research impact through synergistic integration of diverse expertise and methodologies:
Structural Biology and Medicinal Chemistry Integration:
Structural biologists provide high-resolution structures of PPCS in various states
Medicinal chemists leverage these structures for rational drug design
Computational chemists conduct virtual screening and molecular dynamics simulations
Collaborative outcome: Structure-based development of specific PPCS modulators with therapeutic potential
Clinical Genetics and Biochemistry Synergy:
Clinical geneticists identify novel PPCS variants in patient populations
Biochemists characterize functional consequences of these variants
Bioinformaticians develop predictive algorithms for variant pathogenicity
Collaborative outcome: Improved diagnostic accuracy and personalized treatment approaches
Systems Biology and Metabolic Research Combination:
Systems biologists model CoA metabolism in broader metabolic networks
Metabolic researchers experimentally validate model predictions
Bioinformaticians integrate multi-omics data to refine models
Collaborative outcome: Comprehensive understanding of PPCS's role in metabolic regulation
Neuroscience and Cardiology Cross-disciplinary Approach:
Neurologists characterize PPCS-related neurological manifestations
Cardiologists document cardiac phenotypes in PPCS variant carriers
Basic scientists investigate tissue-specific PPCS functions
Collaborative outcome: Mechanistic understanding of tissue-specific disease manifestations
Data Science and Clinical Research Application:
Data scientists apply frameworks like PCS to ensure research reproducibility
Clinical researchers collect standardized patient data using agreed protocols
Biostatisticians develop appropriate analysis methods for heterogeneous data
Collaborative outcome: More robust, generalizable findings with clinical relevance
Pharmaceutical Industry and Academic Partnership:
Academic researchers conduct fundamental PPCS biology studies
Industry partners provide screening capabilities and drug development expertise
Regulatory experts guide translational pathway development
Collaborative outcome: Accelerated development of PPCS-targeted therapeutics
Patient Advocacy and Research Coordination:
Patient advocacy groups provide critical perspectives on research priorities
Researchers ensure studies address clinically relevant questions
Clinicians bridge research findings to patient care
Collaborative outcome: More patient-centered research with improved clinical relevance
Phosphopantothenoylcysteine Synthetase (PPCS) is an enzyme that plays a crucial role in the biosynthesis of coenzyme A (CoA) from pantothenic acid (vitamin B5). This enzyme is essential for various metabolic pathways in both prokaryotes and eukaryotes. The human recombinant form of PPCS is particularly significant for research and therapeutic applications.
The biosynthesis of CoA is a vital process for cellular metabolism. CoA is a cofactor that is involved in various biochemical reactions, including the synthesis and oxidation of fatty acids, and the metabolism of carbohydrates and amino acids. The role of PPCS in this pathway underscores its importance in maintaining cellular function and energy production .
Mutations in the PPCS gene have been associated with certain diseases, such as dilated cardiomyopathy (CMD2C) and familial isolated dilated cardiomyopathy . Understanding the function and regulation of PPCS can provide insights into these conditions and potentially lead to the development of targeted therapies.