Recombinant Putative S-adenosyl-L-methionine-dependent methyltransferase MAP_2076c (MAP_2076c)

Shipped with Ice Packs
In Stock

Description

Introduction

Recombinant Putative S-adenosyl-L-methionine-dependent methyltransferase MAP_2076c (MAP_2076c) is a protein that functions as a methyltransferase, utilizing S-adenosylmethionine (SAM) as a cofactor. Methyltransferases are enzymes that catalyze the transfer of a methyl group from a donor (SAM) to an acceptor molecule. This modification is crucial in various biological processes, including DNA methylation, protein methylation, and the synthesis of various metabolites .

Functional Aspects

MAP_2076c is involved in the transfer of methyl groups, a common biochemical modification with a wide array of effects on protein function and regulation . S-adenosylmethionine (SAM) serves as the methyl donor in these reactions . Methylation can alter enzyme activity, protein-protein interactions, and cellular signaling pathways.

S-Adenosylmethionine (SAM) Biosynthesis

S-adenosylmethionine (SAM) is synthesized from L-methionine and ATP by methionine adenosyltransferase (MAT) . Mammals have isoenzymes like MAT1A (liver-specific) and MAT2A (ubiquitously expressed), with MAT2B regulating MAT2A activity .

Methyl Group Introduction

Introduction of a methyl group can significantly impact the stability and activity of molecules. For example, the addition of a methyl group at the 6-position of a pyrido[3,4-d]pyrimidine core improved human liver microsome (HLM) stability, likely by blocking the preferred pharmacophore for P450 recognition .

Tables in Data Presentation

Tables are used to organize complex data, allowing readers to quickly understand results . Effective tables should have clear titles and descriptive column heads. Tables are best used to present precise numerical values and compare data with shared characteristics .

Table 1: Guidelines for Data Presentation

Use a TableUse a FigureUse Text
To show many and precise numerical values and other specific data in a small spaceTo show trends, patterns, and relationships across and between datasetsWhen you don't have extensive data to present
To compare and contrast data values with several shared characteristicsTo summarize research resultsWhen a table would have two or fewer columns
To show the presence or absence of specific characteristicsTo present a visual explanation of a sequence of events, procedures, or characteristicsWhen the data is irrelevant to the main study findings

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference during order placement for customized preparation.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to sediment the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, provided as a reference for your use.
Shelf Life
Shelf life depends on several factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during the manufacturing process.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
MAP_2076c; Putative S-adenosyl-L-methionine-dependent methyltransferase MAP_2076c; EC 2.1.1.-
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-310
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Mycobacterium paratuberculosis (strain ATCC BAA-968 / K-10)
Target Names
MAP_2076c
Target Protein Sequence
MPRTDNDTWD LSTSVGATAT MVAAARAIAT NADNPLIEDR FAEPLVRAVG VDFFTRWVTG DLVAADVDDH DSGWKLEHMP VAMAARTRFF DSFFQAATQA GIRQAVILAS GLDARAYRLA WPAGTTVFEI DQPQVIEFKT ATLAKLGATP QATLRTVAVD LRDDWPKALV EAGFDKGQPT AWIAEGLFGY LPPEAQDRLL DNITALSADG SRLACEAIPD MSEVDTEKAQ EMMRRATAKW REHGFDLEFG DLGYQGERND VAEYLDGLGW RSVGVPMSQL LADAGLEAIP QTNDSVSVAD TIYYSSVLAK
Uniprot No.

Target Background

Function
Exhibits S-adenosyl-L-methionine-dependent methyltransferase activity.
Database Links
Protein Families
UPF0677 family

Q&A

What is the general function of S-adenosyl-L-methionine-dependent methyltransferases?

S-adenosyl-L-methionine (AdoMet/SAM)-dependent methyltransferases transfer methyl groups from AdoMet to nitrogen, carbon, or oxygen compounds in a wide variety of substrates. This methylation can modify DNA, RNA, proteins, lipids, and small molecules. In bacterial systems, these modifications serve several critical functions:

  • Distinguishing between self and non-self DNA

  • Directing postreplicative mismatch repair

  • Controlling DNA replication and cell cycle

  • Modifying protein function and activity

  • Regulating gene expression

The addition of methyl groups to these molecules provides epigenetic information that can alter the targeting and timing of gene expression and the activity of certain enzymes .

How are bacterial methyltransferases classified structurally and functionally?

Bacterial methyltransferases can be classified based on several characteristics:

Classification BasisCategoriesExamplesKey Features
Substrate SpecificityDNA MTasesDam, DcmModify specific DNA sequences
Protein MTasesPrmC, MAP_2076c (putative)Modify specific amino acid residues
RNA MTasesRlmN, RsmAModify specific RNA positions
Methylation TargetN-MTasesPrmCMethylate nitrogen atoms
C-MTasesRv2067cMethylate carbon atoms
O-MTasesCbiLMethylate oxygen atoms
Structural MotifsClass IPrmCRossmann fold for SAM binding
Class II-VVariousAlternative SAM-binding folds

Although specific functional information about MAP_2076c is limited, it likely belongs to the protein MTase category based on sequence homology with other characterized bacterial methyltransferases .

What experimental approaches are used to identify the substrates of novel methyltransferases?

Identifying substrates for novel methyltransferases requires a systematic approach combining multiple techniques:

  • Bioinformatic prediction: Sequence analysis and structural homology modeling can provide initial hypotheses about potential substrates.

  • Pull-down assays: Using tagged recombinant methyltransferase as bait to identify interacting partners, as demonstrated with Rv2067c which was initially identified in pulldown experiments with histone-like protein MtHU .

  • In vitro methylation assays: Testing potential substrates using purified recombinant methyltransferase with tritiated SAM as a methyl donor, followed by detection through autoradiography or scintillation counting .

  • Mass spectrometry analysis: To identify specific methylation sites, as demonstrated for Rv2067c where MS/MS identified H3K79 as the target of methylation with a mass shift of 42 Da indicating trimethylation .

  • Antibody-based detection: Using methylation-specific antibodies in dot blots or Western blots to confirm methylation events .

  • Comparative analysis: Testing candidate substrates based on known targets of homologous enzymes, as seen when recombinant PrmC from C. trachomatis was tested against release factors based on E. coli PrmC function .

What expression systems are optimal for recombinant MAP_2076c production?

The choice of expression system for MAP_2076c should be based on experimental goals and protein characteristics:

Expression SystemAdvantagesDisadvantagesConsiderations for MAP_2076c
E. coliHigh yield, rapid growth, well-established protocolsLack of mycobacterial post-translational modificationsUse vectors like pQE-80L as demonstrated for C. trachomatis PrmC
Mycobacterial hostsAuthentic post-translational modifications, proper foldingSlower growth, lower yieldsRecommended for functional studies in native-like environment
Cell-free systemsAvoid toxicity issues, rapid productionHigher cost, limited scaleUseful if MAP_2076c expression proves toxic to cells
Mammalian cellsHost-relevant modificationsComplex, expensive, lower yieldConsider for host-pathogen interaction studies

When expressing potentially toxic proteins like methyltransferases, it's important to note that even low levels of expression may be sufficient for functional studies, as demonstrated with C. trachomatis PrmC where undetectable levels by SDS-PAGE were sufficient for complementation .

What purification strategies maximize yield and activity of recombinant methyltransferases?

Effective purification of active methyltransferases requires careful consideration of several factors:

  • Affinity tags: Histidine tags allow for simplified purification using nickel affinity chromatography, but consider tag position (N- or C-terminal) to avoid interference with activity.

  • Buffer optimization: Include SAM or SAH in purification buffers to stabilize the enzyme's active site, and add reducing agents to prevent oxidation of catalytic residues.

  • Column chromatography sequence:

    • Initial capture: Affinity chromatography (His-tag, GST, etc.)

    • Intermediate purification: Ion exchange chromatography

    • Polishing: Size exclusion chromatography to remove aggregates

  • Activity preservation:

    • Add glycerol (10-20%) to prevent freezing damage

    • Include protease inhibitors to prevent degradation

    • Minimize freeze-thaw cycles by storing in small aliquots

  • Quality control: Verify enzyme activity at each purification step and assess homogeneity by SDS-PAGE and dynamic light scattering.

For methyltransferases like MAP_2076c, maintaining the native conformation of the SAM-binding domain is critical for preserving enzymatic activity.

How can complementation assays be designed to verify MAP_2076c function?

Complementation studies provide powerful evidence for functional conservation and can be designed as follows:

  • Heterologous complementation in E. coli:

    • Utilize an E. coli strain with a knockout of a related methyltransferase gene (e.g., prmC)

    • Transform with a plasmid expressing MAP_2076c

    • Assess rescue of growth defects or other phenotypes

This approach was successfully used for C. trachomatis PrmC, which complemented an E. coli prmC knockout strain, demonstrating functional conservation despite evolutionary distance .

  • Homologous recombination in mycobacteria:

    • Generate a MAP_2076c knockout in M. avium or a related mycobacterial species

    • Create complementation strains with:

      • Wild-type MAP_2076c (positive control)

      • Catalytically inactive mutant (negative control)

      • Chimeric constructs to map functional domains

  • Controls and measurements:

    • Growth curves to assess complementation of growth defects

    • Specific phenotypic assays relevant to the putative function

    • Molecular readouts such as substrate methylation status

Successful complementation provides strong evidence for functional homology and can reveal the minimum expression level required for activity, as seen with C. trachomatis PrmC where complementation occurred even when recombinant protein was undetectable by SDS-PAGE .

What in vitro assays accurately measure methyltransferase activity of MAP_2076c?

Several complementary approaches can quantitatively measure methyltransferase activity:

  • Radiometric assays:

    • Use tritiated SAM ([³H]-SAM) as the methyl donor

    • Incubate with purified MAP_2076c and potential substrate

    • Measure incorporation of radioactive methyl groups by scintillation counting

    • This approach was used successfully for Rv2067c methylation activity on histone H3

  • Mass spectrometry-based assays:

    • Incubate MAP_2076c with substrate and SAM

    • Digest with appropriate protease for peptide analysis

    • Detect mass shifts corresponding to methylation (e.g., +14 Da for monomethylation)

    • Quantify levels of methylated vs. unmethylated peptides

    • This method identified the H3K79 trimethylation (+42 Da) by Rv2067c

  • Antibody-based detection:

    • Develop or obtain antibodies specific to methylated substrates

    • Perform dot blot or Western blot analysis after methylation reaction

    • Quantify signal intensity to measure methylation levels

    • This approach confirmed H3K79 trimethylation by Rv2067c

  • Enzyme-coupled assays:

    • Measure SAH production using coupled enzymatic reactions

    • Monitor spectrophotometrically for real-time kinetic analysis

A combination of these methods provides robust validation of methyltransferase activity and substrate specificity.

How can structural studies inform the catalytic mechanism of MAP_2076c?

Structural analysis provides critical insights into methyltransferase mechanisms:

  • X-ray crystallography:

    • Determine the three-dimensional structure at atomic resolution

    • Co-crystallize with SAM/SAH to visualize cofactor binding

    • Co-crystallize with substrate to identify binding interface

    • Analyze active site architecture for catalytic residues

  • Comparative structural analysis:

    • Compare with structures of related methyltransferases

    • Identify conserved structural elements across methyltransferase families

    • Map sequence conservation onto structural models

  • Structure-guided mutagenesis:

    • Design mutations of predicted catalytic residues

    • Create SAM-binding mutants (e.g., RxR mutations as used for Rv2067c )

    • Test effects on activity using assays described above

  • Molecular dynamics simulations:

    • Model conformational changes during catalysis

    • Predict effects of mutations on protein stability and activity

    • Simulate substrate binding and product release

Structural studies of Rv2067c and DOT1L revealed the basis for their differential activity on H3K79 in free versus nucleosomal contexts, demonstrating how structural information can explain functional specificity .

What computational approaches help predict MAP_2076c substrate specificity?

Computational methods offer valuable tools for predicting substrate specificity:

Computational ApproachApplicationToolsOutput
Homology modelingPredict 3D structureAlphaFold, SWISS-MODELStructural model of MAP_2076c
Molecular dockingPredict substrate bindingAutoDock, HADDOCKBinding poses, interaction energies
Sequence motif analysisIdentify conserved catalytic residuesMEME, PROSITEConserved sequence patterns
Phylogenetic analysisIdentify functional relationshipsMEGA, PhyMLEvolutionary relationships with characterized MTases
Molecular dynamicsSimulate enzyme-substrate interactionsGROMACS, NAMDDynamics of substrate recognition
Machine learningPredict substrate specificityTensorFlow, PyTorchProbability scores for potential substrates

These approaches can generate testable hypotheses about MAP_2076c function based on similarity to well-characterized methyltransferases like PrmC and Rv2067c .

How might MAP_2076c contribute to Mycobacterium avium pathogenesis?

Based on findings with related mycobacterial methyltransferases, MAP_2076c may play important roles in pathogenesis:

  • Host epigenetic manipulation:

    • Mycobacterium tuberculosis MTase Rv2067c secretes into host macrophages

    • Trimethylates histone H3K79 in non-nucleosomal context

    • Downregulates host MTase DOT1L

    • Inhibits caspase-8-dependent apoptosis

    • Enhances RIPK3-mediated necrosis

    • Increases pathogen survival and virulence

By analogy, MAP_2076c might similarly manipulate host processes through methylation of key host proteins, potentially explaining aspects of M. avium persistence in host cells.

  • Bacterial stress adaptation:

    • Methylation of bacterial proteins may enhance survival under stress conditions

    • Modification of translation machinery components (like release factors by PrmC )

    • Regulation of gene expression through methylation of transcription factors

  • Immune evasion:

    • Modification of bacterial surface antigens

    • Manipulation of host immune signaling pathways

    • Alteration of host cell death mechanisms

Experimental approaches to investigate these possibilities include:

  • Comparing virulence of wild-type and MAP_2076c knockout strains in cellular and animal models

  • Analyzing methylation targets during infection

  • Examining host transcriptional responses to MAP_2076c exposure

What host factors might interact with or be modified by MAP_2076c during infection?

Based on studies of related methyltransferases, MAP_2076c might interact with various host factors:

Potential Host TargetEvidence from Related MTasesFunctional ConsequenceDetection Method
Histone proteinsRv2067c methylates H3K79 Altered gene expressionMS/MS, ChIP-seq
Cell death regulatorsRv2067c inhibits caspase-8-dependent apoptosis Enhanced bacterial survivalApoptosis assays, co-IP
Inflammatory mediatorsRv2067c enhances SESTRIN3, NLRC3, TMTC1 expression Suppressed inflammationRNA-seq, qRT-PCR
Pattern recognition receptorsHypothetical based on immune evasion strategiesImpaired pathogen detectionProtein methylation assays
Cytoskeletal componentsHypothetical based on intracellular lifestyleAltered bacterial traffickingImmunofluorescence microscopy

To identify such interactions:

  • Perform pull-down assays with recombinant MAP_2076c using host cell lysates

  • Conduct comparative proteomics between cells infected with wild-type vs. ΔMAP_2076c

  • Use proximity labeling techniques to identify proteins in close association with MAP_2076c during infection

How can CRISPR-Cas9 technology facilitate MAP_2076c functional studies?

CRISPR-Cas9 technology offers powerful approaches for MAP_2076c research:

  • Gene knockout strategies:

    • Generate precise MAP_2076c deletions in M. avium

    • Create scarless mutations to minimize polar effects

    • Develop conditional knockouts for essential genes

  • Domain mapping:

    • Introduce specific mutations in catalytic residues

    • Create truncated versions to identify minimal functional domains

    • Modify potential regulatory regions

  • Reporter systems:

    • Knock-in fluorescent tags for localization studies

    • Create activity-dependent reporters to monitor methylation

    • Develop biosensors for real-time activity measurement

  • Host-pathogen studies:

    • Modify host genes encoding potential MAP_2076c substrates

    • Generate substrate-resistant variants (e.g., mutate target residues)

    • Create reporter cell lines to detect methylation events

  • High-throughput screening:

    • Generate CRISPR libraries to identify host factors involved in MAP_2076c function

    • Screen for synthetic lethal interactions with MAP_2076c in bacterial systems

CRISPR-based approaches provide unprecedented precision in genetic manipulation, enabling detailed dissection of MAP_2076c function in both bacterial and host contexts.

What are the most promising inhibitor development strategies for MAP_2076c?

Developing specific inhibitors for MAP_2076c could have both research and therapeutic applications:

  • Structure-based design approaches:

    • Virtual screening against the SAM-binding pocket

    • Fragment-based drug discovery targeting substrate binding site

    • Design of bisubstrate analogs bridging SAM and substrate binding sites

  • High-throughput screening strategies:

    • Biochemical assays measuring methyltransferase activity

    • Cell-based assays monitoring infection outcomes

    • Displacement assays for SAM binding

  • Repurposing existing methyltransferase inhibitors:

    • Testing known DOT1L inhibitors against MAP_2076c

    • Evaluating broad-spectrum methyltransferase inhibitors

    • Modifying existing compounds for increased specificity

  • Targeting unique structural features:

    • Identifying allosteric sites specific to MAP_2076c

    • Developing compounds that exploit differences from host methyltransferases

    • Designing inhibitors that block potential secretion mechanisms

  • Combination approaches:

    • Pairing with conventional antimycobacterials

    • Targeting multiple mycobacterial methyltransferases simultaneously

    • Combining with host-directed therapies

The development of selective inhibitors would not only provide research tools but could potentially lead to novel therapeutic strategies against M. avium infections.

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 2025 TheBiotek. All Rights Reserved.