PCMT1 Human

Protein-L-Isoaspartate O-Methyltransferase Human Recombinant
Shipped with Ice Packs
In Stock

Description

PCMT1 Recombinant Human produced in E.Coli is a single, non-glycosylated polypeptide chain containing 263 amino acids (1-227 a.a.) and having a molecular mass of 28.8 kDa. The PCMT1 is fused to a 36 amino acids His-Tag at N-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction
The enzyme PCMT1 catalyzes the methyl esterification of L-isoaspartyl and D-aspartyl residues found in peptides and proteins. These residues are a result of the spontaneous decomposition of normal L-aspartyl and L-asparaginyl residues. PCMT1 plays a crucial role in the repair or degradation of damaged proteins.
Description
Recombinant Human PCMT1, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 263 amino acids (with a sequence spanning from amino acid 1 to 227) and possessing a molecular mass of 28.8 kDa. A 36 amino acid His-Tag is fused to the N-terminus of PCMT1. The protein undergoes purification using proprietary chromatographic techniques.
Physical Appearance
A clear and sterile solution.
Formulation
PCMT1 Human solution is provided in a buffer containing 20mM Tris-HCl at pH 8, 0.1M NaCl, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to store the product frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advisable for long-term storage. It's essential to avoid repeated freeze-thaw cycles.
Purity
The purity of the PCMT1 protein is greater than 95%, as determined by SDS-PAGE analysis.
Synonyms
EC 2.1.1.77, PIMT, PCMT1, Protein-beta-aspartate methyltransferase, Protein L-isoaspartyl/D-aspartyl methyltransferase, L-isoaspartyl protein carboxyl methyltransferase.
Source
Escherichia Coli.
Amino Acid Sequence
RGSHHHHHH GMASMTGGQQ MGRDLYDDDD KDRWGSMAWK SGGASHSELI HNLRKNGIIK TDKVFEVMLA TDRSHYAKCN PYMDSPQSIG FQATISAPHM HAYALELLFD QLHEGAKALD VGSGSGILTA CFARMVGCTG KVIGIDHIKE LVDDSINNVR KDDPTLLSSG RVQLVVGDGR MGYAEEAPYD AIHVGAAAPV VPQALIDQLK PGGRLILPVG PAGGNQMLEQ YDKLQDGSIK MKPLMGVIYV PLTDKEKQWS RWK.

Q&A

What is the primary function of PCMT1 in human cells?

PCMT1 (Protein-L-isoaspartate (D-aspartate) O-methyltransferase) is an S-adenosylmethionine-dependent enzyme that catalyzes the methyl esterification of L-isoaspartyl and D-aspartyl residues in damaged proteins . These abnormal residues form through spontaneous isomerization and racemization of L-aspartyl and L-asparaginyl residues during protein aging . Through this methylation process, PCMT1 initiates the repair of damaged proteins, effectively restoring their original structure and function . The enzyme acts on numerous substrates including EIF4EBP2, microtubule-associated protein 2, calreticulin, clathrin light chains, and several synucleins .

Where is PCMT1 predominantly expressed in human tissues?

While PCMT1 is expressed in all human tissues, expression levels vary significantly. The highest expression is found in neural tissues, particularly the brain . This distribution pattern aligns with its critical role in protecting neural cells against protein damage and maintaining neurological function. In pathological states such as breast cancer, PCMT1 expression patterns become altered, with significant upregulation observed in human breast ductal carcinoma cell lines like HCC1500, HCC1419, and EFM19, while expression remains lower in other breast cancer cell lines such as HCC202 .

What genomic location and structural features characterize the PCMT1 gene?

The PCMT1 gene is located on chromosome 6 in humans . The gene encodes a protein repair enzyme that contains specific binding motifs for L-isoaspartate and S-adenosyl-L-methionine (SAM), which serves as the methyl donor in the repair reaction . The protein contains a SAM-binding motif that is crucial for its protective function against Bax-induced apoptosis in neural cells . This methyltransferase belongs to type II protein carboxyl methyltransferases, distinguished by their substrate specificity for damaged aspartyl residues .

What methods are most effective for measuring PCMT1 expression in clinical samples?

For PCMT1 expression analysis in clinical contexts, researchers typically employ a multi-platform approach. RNA-sequencing data analysis, as utilized in TCGA and GTEx projects, provides robust quantification of PCMT1 transcript levels across various cancer types . For protein-level validation, immunohistochemistry techniques through the Human Protein Atlas database offer visual confirmation of expression patterns in tissue samples .

For higher sensitivity in expression analysis, quantitative PCR remains valuable, particularly when working with limited clinical material. When analyzing PCMT1's relationship with clinical parameters, computational tools like bc-GenExMiner provide powerful platforms for correlating expression with pathological features . For comprehensive methyltransferase activity assessment, enzymatic assays measuring the transfer of methyl groups from S-adenosyl-L-methionine to protein substrates containing L-isoaspartyl residues can provide functional insights beyond mere expression levels.

How can PCMT1 knockout models be effectively developed and validated?

Development of PCMT1 knockout models requires careful consideration of the severe phenotype observed in complete knockout. Since Pcmt1 null (pcmt1-/-) mice die approximately 42 days after birth due to progressive epileptic seizures , researchers should consider:

  • Conditional knockout systems using Cre-loxP technology to restrict PCMT1 deletion to specific tissues or developmental stages

  • Inducible knockout systems that allow temporal control of gene silencing

  • Hypomorphic models that reduce rather than eliminate PCMT1 expression

Validation should include:

  • Genomic verification through PCR and sequencing

  • Transcript analysis via RT-qPCR

  • Protein depletion confirmation through Western blotting

  • Functional validation by measuring accumulation of L-isoaspartyl residues in target tissues

  • Phenotypic characterization focusing on neurological parameters, given the high PCMT1 expression in neural tissues and seizure phenotype in null mice

What are the optimal approaches for studying PCMT1's enzymatic activity in vitro?

Studying PCMT1's enzymatic activity requires specialized techniques focused on methyltransferase function:

  • Recombinant protein expression: Full-length PCMT1 should be expressed with appropriate tags (e.g., N-terminal 6xHis-tags) in expression vectors like pMAPle4, which allow for purification while maintaining enzymatic function .

  • Substrate preparation: Utilize artificially aged proteins containing L-isoaspartyl residues or synthetic peptides with incorporated L-isoaspartyl residues as substrates.

  • Activity assays: Measure methyltransferase activity through:

    • Radiometric assays using [³H]-S-adenosyl-L-methionine to quantify methyl transfer

    • HPLC-based methods to detect formation of methylated products

    • Coupled enzyme assays that monitor S-adenosylhomocysteine (SAH) production

  • Kinetic analysis: Determine enzyme kinetics parameters (Km, Vmax) for various substrates to understand substrate preferences and catalytic efficiency.

  • Inhibition studies: Evaluate potential inhibitors by measuring their effects on PCMT1 activity using IC50 and Ki determinations.

These approaches allow comprehensive characterization of PCMT1's enzymatic properties, substrate specificity, and potential modulation by small molecules.

How does PCMT1 expression correlate with cancer prognosis across different tumor types?

PCMT1 demonstrates significant prognostic value across multiple cancer types, with particularly strong evidence in breast cancer. Pan-cancer analysis reveals that PCMT1 is aberrantly expressed in numerous tumors, with significant upregulation observed in breast cancer (BRCA), cervical squamous cell carcinoma (CESC), cholangiocarcinoma (CHOL), colon adenocarcinoma (COAD), and several other cancer types .

In breast cancer specifically, high PCMT1 expression correlates with poorer patient outcomes. Survival analysis demonstrates that elevated PCMT1 expression is significantly associated with:

The prognostic value of PCMT1 extends beyond breast cancer to other malignancies. It has been identified as a predictive marker for poor prognosis in lung adenocarcinoma and serves as an unfavorable prognostic biomarker for bladder cancer . The consistency of these findings across multiple cancer types suggests PCMT1 may function as a pan-cancer prognostic indicator, with its overexpression generally associated with more aggressive disease phenotypes.

What mechanisms underlie PCMT1's role in cancer progression and metastasis?

PCMT1 contributes to cancer progression through several molecular mechanisms:

  • Epithelial-mesenchymal transition (EMT) regulation: PCMT1 participates in cancer cell migration and invasion by modulating EMT-related genes, thereby functioning as an oncogene in bladder cancer .

  • Migration and invasion promotion: Research demonstrates that PCMT1 enhances the migratory and invasive capabilities of human glioblastoma cell lines (U-87 MG and U-251 MG) and plays a crucial role in glioblastoma growth .

  • Immune microenvironment modulation: PCMT1 expression significantly correlates with immune cell infiltration in various cancers, particularly breast cancer, potentially affecting tumor immunosurveillance and response to immunotherapy .

  • Molecular pathway interactions: While specific pathways remain under investigation, PCMT1's protein repair function may preserve the function of oncogenic proteins that would otherwise be degraded due to isoaspartyl damage, thereby supporting cancer cell survival and proliferation.

These mechanisms collectively contribute to PCMT1's role in cancer progression, though additional research is needed to fully elucidate the specific molecular pathways through which PCMT1 drives metastatic processes.

What is the relationship between PCMT1 expression and tumor immune microenvironment?

PCMT1 exhibits significant associations with the tumor immune microenvironment across multiple cancer types:

  • Immune cell infiltration: Comprehensive analysis reveals strong correlations between PCMT1 expression and levels of immune cell infiltrates . In breast cancer specifically, PCMT1 expression significantly relates to the abundance of immune infiltration .

  • Immune checkpoint correlation: PCMT1 expression correlates with eight key immune checkpoints, including CD274, SIGLEC15, CTLA-4, HAVCR2, PDCD1, LAG3, PDCD1LG2, and TIGIT . These checkpoints are critical regulators of anti-tumor immune responses and targets for immunotherapy.

  • Immunotherapy response predictors: PCMT1 expression correlates with tumor mutation burden (TMB) and microsatellite instability (MSI) , both established predictors of response to immune checkpoint inhibitor therapies (ICIs).

  • Immune cell subset analysis: Using single sample Gene Set Enrichment Analysis (ssGSEA), relationships between PCMT1 expression and 24 specific immune cell types have been characterized in breast cancer, providing detailed insights into how PCMT1 may shape the immune landscape .

These findings suggest PCMT1 may influence cancer progression partially through modulation of the tumor immune microenvironment, with potential implications for immunotherapy response prediction and treatment strategies.

How does post-translational modification affect PCMT1 activity and substrate specificity?

PCMT1 function itself can be regulated through post-translational modifications, though this area requires further research. Current understanding suggests:

  • Phosphorylation: PCMT1 contains potential phosphorylation sites that may modulate its enzymatic activity, subcellular localization, or substrate recognition. Kinase-mediated regulation could provide rapid control of PCMT1 function in response to cellular stressors.

  • Oxidative modifications: As a protein involved in protein damage repair, PCMT1 itself may be susceptible to oxidative damage, potentially creating feedback regulation where its own activity diminishes under oxidative stress conditions.

  • Compartmentalization effects: Post-translational modifications may affect PCMT1's subcellular distribution, potentially regulating its access to different substrates in various cellular compartments.

For experimental investigation, researchers should consider:

  • Phosphoproteomic analysis to identify modification sites

  • Site-directed mutagenesis to create modification-mimetic or modification-resistant PCMT1 variants

  • In vitro activity assays comparing native and modified PCMT1 against diverse substrates

  • Subcellular fractionation studies to determine how modifications affect PCMT1 localization

Such investigations would provide crucial insights into the regulation of this important protein repair enzyme.

What is the interplay between PCMT1 and one-carbon metabolism in cancer cells?

PCMT1 is intricately connected to one-carbon metabolism as an S-adenosyl-L-methionine (SAM)-dependent methyltransferase, creating a potentially significant intersection with cancer metabolism:

  • SAM consumption: PCMT1 activity requires SAM as a methyl donor, producing S-adenosylhomocysteine (SAH) as a byproduct. This places PCMT1 within the methionine cycle component of one-carbon metabolism.

  • Metabolic alterations: Studies in Pcmt1 null (pcmt1-/-) mice revealed significantly higher SAM levels and lower SAH levels in brain tissue compared to wild-type littermates , suggesting PCMT1 influences the SAM:SAH ratio and potentially broader aspects of one-carbon metabolism.

  • Folate pathway connections: Upregulation of Pcmt1 was observed when the normal phenotype was rescued with folic acid supplementation in Folr1 (folate-binding protein 1) nullizygous mice , indicating potential crosstalk between PCMT1 and folate metabolism.

  • Cancer cell metabolic vulnerabilities: In rapidly proliferating cancer cells with high protein synthesis rates, PCMT1-mediated protein repair may represent a significant consumer of SAM molecules, potentially creating targetable metabolic dependencies.

This interplay suggests PCMT1 inhibition could potentially disrupt one-carbon metabolism in cancer cells, representing a novel therapeutic angle worth investigating through metabolomic profiling and isotope tracing experiments.

How might PCMT1 contribute to treatment resistance in cancer therapies?

PCMT1 may contribute to treatment resistance through several potential mechanisms:

  • Protein damage repair: Many cancer therapies, particularly radiation and some chemotherapeutics, induce oxidative stress that can damage proteins through racemization and isomerization. PCMT1's protein repair function may counteract this damage, helping cancer cells maintain proteostasis despite treatment-induced stress.

  • Immune checkpoint modulation: Given PCMT1's correlation with immune checkpoints like CTLA-4, PD-1 (PDCD1), and PD-L1 (CD274) , it may influence response to immune checkpoint inhibitor therapies. High PCMT1 expression could potentially contribute to an immunosuppressive tumor microenvironment that limits immunotherapy efficacy.

  • Survival pathway maintenance: By repairing damaged signaling proteins involved in proliferation and survival pathways, PCMT1 may help maintain oncogenic signaling despite therapeutic intervention.

  • Association with prognostic factors: PCMT1's correlation with tumor mutation burden (TMB) and microsatellite instability (MSI) links it to factors known to affect treatment response across multiple modalities.

Investigation approaches could include:

  • Combining PCMT1 inhibition with standard therapies in resistant cell lines

  • Analyzing PCMT1 expression in matched pre-treatment and post-relapse patient samples

  • Correlating PCMT1 expression with treatment outcomes in clinical cohorts

  • Screening for PCMT1-dependent proteins involved in known resistance mechanisms

These investigations could potentially identify PCMT1 as a therapeutic target to overcome treatment resistance.

How can PCMT1 expression be effectively used as a prognostic biomarker in clinical settings?

Implementing PCMT1 as a clinical prognostic biomarker requires standardized approaches:

These steps would facilitate translation of PCMT1's demonstrated prognostic value from research findings to clinical utility.

What strategies might be effective for targeting PCMT1 therapeutically in cancer?

Therapeutic targeting of PCMT1 could employ several strategic approaches:

  • Direct enzyme inhibition: Development of small molecule inhibitors that:

    • Compete with SAM for binding to PCMT1

    • Occupy the substrate binding pocket

    • Allosterically modify PCMT1 structure to impair catalytic function

  • Transcriptional regulation: Approaches to reduce PCMT1 expression:

    • Targeted antisense oligonucleotides or siRNAs

    • Identification of transcription factors driving PCMT1 expression

    • Epigenetic modifiers affecting PCMT1 promoter accessibility

  • Protein degradation: Inducing PCMT1 protein elimination via:

    • Proteolysis targeting chimeras (PROTACs) specific to PCMT1

    • Autophagy-inducing compounds that enhance PCMT1 turnover

    • Destabilization of PCMT1 through disruption of stabilizing interactions

  • Synthetic lethality approaches: Identify cellular contexts where PCMT1 inhibition would be selectively lethal:

    • High oxidative stress environments where protein damage is elevated

    • Tumors with specific mutational profiles that increase dependency on PCMT1

    • Combinations with therapies that increase isoaspartyl formation

  • Immune modulation: Given PCMT1's correlation with immune infiltration and checkpoints:

    • Combine PCMT1 inhibition with immune checkpoint blockade

    • Target PCMT1 to potentially enhance tumor antigen presentation

    • Modify tumor microenvironment immune composition

Development of these strategies should include consideration of potential toxicities, particularly neurological effects, given PCMT1's high expression and important function in neural tissues.

What are the potential applications of PCMT1 research in neurodegenerative disorders?

PCMT1's protein repair function has significant implications for neurodegenerative disorders:

  • Neuroprotective mechanisms: PCMT1 protects certain neural cells against Bax-induced apoptosis through an SAM-binding motif-dependent mechanism . This neuroprotective function suggests potential applications in:

    • Preventing neuronal loss in neurodegenerative conditions

    • Preserving neural function during aging

    • Protecting against excitotoxicity-induced neuronal damage

  • Protein aggregation and misfolding: Many neurodegenerative disorders involve protein aggregation. PCMT1's role in:

    • Repairing α-synuclein (implicated in Parkinson's disease)

    • Potentially affecting tau protein processing (Alzheimer's disease)

    • Maintaining proper protein conformation in neural tissues

    Makes it relevant to fundamental disease mechanisms.

  • Epilepsy connections: The observation that PCMT1 null mice die from progressive epileptic seizures links PCMT1 to neuronal excitability regulation. PCMT1 is associated with:

    • Autosomal dominant epilepsy with auditory features

    • Benign familial infantile epilepsy

    • Progressive myoclonic epilepsy type 6

    • Developmental and epileptic encephalopathy

  • Therapeutic approaches:

    • PCMT1 enhancement strategies for protein repair in aging brains

    • Targeted delivery of PCMT1 to affected neural populations

    • Small molecules that mimic or enhance PCMT1 activity

Future research directions should include examining PCMT1 expression and activity in neurodegenerative disease tissue samples, evaluating genetic variants in neurodegenerative disease cohorts, and developing neural-specific PCMT1 modulation strategies.

What novel technologies could advance understanding of PCMT1's substrates and interactome?

Emerging technologies offer promising approaches for comprehensive characterization of PCMT1's substrates and interaction network:

  • Proteomic identification of PCMT1 substrates:

    • PIMT-catalyzed O-methylesterification coupled with mass spectrometry to identify isoaspartyl-containing substrates

    • SILAC-based approaches comparing wild-type and PCMT1-knockout cells to identify accumulating damaged proteins

    • Chemical proteomics using modified SAM analogs to tag and identify PCMT1 substrates

  • Interactome mapping technologies:

    • Proximity labeling methods (BioID, APEX) to identify proteins in close spatial proximity to PCMT1

    • Quantitative interaction proteomics using stable isotope labeling

    • Hydrogen-deuterium exchange mass spectrometry to identify dynamic PCMT1 interaction interfaces

    • High-throughput yeast two-hybrid or mammalian two-hybrid screens

  • Structural biology approaches:

    • Cryo-electron microscopy of PCMT1 in complex with substrates

    • Hydrogen-deuterium exchange mass spectrometry to map binding interfaces

    • Cross-linking mass spectrometry to capture transient interactions

  • Systems biology integration:

    • Multi-omics approaches combining transcriptomics, proteomics, and metabolomics in PCMT1-modulated systems

    • Network analysis tools to position PCMT1 within cellular protein quality control networks

    • Machine learning algorithms to predict additional PCMT1 substrates based on identified patterns

These advanced technologies would significantly expand our understanding of PCMT1's biological function and potentially reveal new therapeutic opportunities.

How might single-cell analysis techniques advance understanding of PCMT1 in heterogeneous tumor environments?

Single-cell technologies offer unprecedented potential to unravel PCMT1's role in complex tumor ecosystems:

  • Single-cell RNA sequencing applications:

    • Identify specific cell populations within tumors expressing high PCMT1 levels

    • Correlate PCMT1 expression with cell states (proliferative, invasive, stem-like)

    • Map co-expression networks in PCMT1-high versus PCMT1-low cells

    • Track PCMT1 expression changes during tumor evolution and treatment response

  • Single-cell protein analysis:

    • Mass cytometry (CyTOF) incorporating PCMT1 antibodies alongside immune markers

    • Single-cell Western blotting to quantify PCMT1 protein levels in rare subpopulations

    • Imaging mass cytometry to preserve spatial context while quantifying PCMT1

  • Spatial transcriptomics:

    • Map PCMT1 expression patterns relative to tumor architecture

    • Identify spatial relationships between PCMT1-expressing tumor cells and specific immune cell subsets

    • Correlate PCMT1 expression with invasive fronts or hypoxic regions

  • Multi-modal single-cell analysis:

    • Combined single-cell transcriptomics and proteomics (CITE-seq) to correlate PCMT1 mRNA and protein

    • Integrate single-cell epigenomic data to understand PCMT1 regulation

    • Single-cell metabolomics to link PCMT1 to SAM utilization at cellular level

  • Clinical applications:

    • Analysis of circulating tumor cells for PCMT1 expression

    • Longitudinal sampling to track PCMT1-expressing clones during treatment

    • Patient-derived organoid models analyzed at single-cell resolution

These approaches would provide crucial insights into the heterogeneity of PCMT1 expression and function within tumors, potentially identifying specific cellular contexts where PCMT1 targeting would be most effective.

What gene editing approaches might advance functional studies of PCMT1?

Advanced gene editing technologies offer powerful tools for PCMT1 functional investigation:

  • CRISPR-Cas9 applications:

    • Complete PCMT1 knockout models with careful consideration of developmental timing

    • Conditional knockout systems using Cre-loxP or Tet-regulated CRISPR

    • Knockin of tagged PCMT1 variants for localization and interaction studies

    • Introduction of specific patient-derived mutations to assess functional consequences

    • Base editing to create subtle modifications without double-strand breaks

  • Domain-specific engineering:

    • Precise editing of SAM-binding motifs

    • Modification of substrate recognition regions

    • Creation of catalytically inactive variants (dead methyltransferase)

    • Generation of isoform-specific modifications

  • High-throughput functional genomics:

    • CRISPR screens to identify synthetic lethal interactions with PCMT1

    • CRISPRa/CRISPRi libraries to identify genes affecting PCMT1 expression

    • Pooled CRISPR screens in the context of various stressors to identify condition-specific PCMT1 dependencies

  • Temporal control systems:

    • Optogenetic regulation of PCMT1 expression

    • Chemical-inducible degron tagging for rapid protein depletion

    • CRISPR interference with inducible promoters for temporal expression control

  • In vivo applications:

    • Tissue-specific PCMT1 modulation using AAV-delivered CRISPR systems

    • Creation of humanized mouse models with patient-specific PCMT1 variants

    • In vivo CRISPR screens focusing on PCMT1 pathway components

These gene editing approaches would provide unprecedented control over PCMT1 expression and function, enabling detailed mechanistic studies and potentially identifying new therapeutic strategies targeting PCMT1-dependent processes.

Product Science Overview

Function and Mechanism

PIMT recognizes and catalyzes the repair of damaged L-isoaspartyl and D-aspartyl groups in proteins. These damaged residues often result from spontaneous deamidation and isomerization of asparagine and aspartate residues, which can occur as proteins age . The enzyme transfers methyl groups from S-adenosyl-L-methionine to the alpha side chain carboxyl groups of these damaged residues, forming a methyl ester. This methyl ester then undergoes nonenzymatic reactions to form L-succinimide, which can be hydrolyzed to generate repaired L-aspartyl residues .

Importance in Protein Repair

The repair mechanism facilitated by PIMT is essential for maintaining protein integrity. Without this repair, abnormal aspartyl residues accumulate, leading to the formation of dysfunctional proteins. In mice, the absence of PIMT has been linked to fatal progressive epilepsy, highlighting the enzyme’s critical role in cellular health .

Human Recombinant PIMT

Human recombinant PIMT is produced using recombinant DNA technology, which allows for the expression of the human enzyme in a host organism, such as bacteria or yeast. This recombinant form is used in various research and therapeutic applications to study the enzyme’s function and to develop potential treatments for diseases associated with protein damage.

Clinical Relevance

Mutations in the PCMT1 gene, which encodes PIMT, have been associated with several disorders, including spina bifida and premature ovarian failure . Additionally, the enzyme may play a protective role in the pathogenesis of Alzheimer’s disease, making it a target for therapeutic research .

Structural Insights

The enzyme exists in human cytosol in two forms due to alternative splicing and varies among individuals due to a single polymorphism at residue 119, which can be either valine or isoleucine . This structural variation can influence the enzyme’s activity and stability.

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.