Recombinant Uncharacterized protein in ramA 3'region

Shipped with Ice Packs
In Stock

Description

Overview of the Recombinant Protein

This protein is a recombinant construct comprising the full-length sequence (1–82 amino acids) of an uncharacterized protein located in the 3' region of the ramA locus. Key characteristics include:

PropertyDetails
Source OrganismKlebsiella pneumoniae
Expression HostE. coli
TagN-terminal His-tag
Molecular Weight~9.2 kDa (estimated from 82 aa)
Protein SequenceMKHPLETLLSAAGILLLALLSCLLLPAPSLGLTLAQKLVETFHMMDLNQLYTVLFCLWFL ALGAIEYLVLRWVWRRWFSLER
Purity>90% (SDS-PAGE verified)
FormLyophilized powder

The ramA locus is known in Klebsiella, Enterobacter, and Salmonella spp. for regulating efflux pumps (e.g., acrAB) linked to antibiotic resistance . While this protein’s role remains undefined, its association with ramA suggests potential involvement in bacterial stress responses or regulatory pathways.

Expression and Production

The protein is produced via recombinant expression in E. coli with an N-terminal His-tag for purification. Critical production parameters include:

Expression and Purification

  • Host Strain: Standard E. coli strains, though codon optimization may be required for high yields .

  • Tag Function: His-tag facilitates affinity chromatography, enabling scalable purification.

  • Yield Challenges: E. coli systems may face issues with rare codon usage, solubility, and inclusion body formation. Strains like BL21(DE3) CodonPlus or Rosetta(DE3) could improve production .

Sequence Features

  • Domain Analysis: No annotated domains (e.g., SANT, BTB) are reported for this protein, unlike other uncharacterized proteins (e.g., SANBR in human cells) .

  • Conservation: Alignments with Klebsiella homologs suggest evolutionary conservation within Enterobacteriaceae .

Potential Roles

  • Regulatory Function: Its proximity to ramA implies possible involvement in transcriptional regulation or efflux pump modulation .

  • Stress Response: ramA-regulated genes often mediate antibiotic resistance via efflux pumps; this protein may interact with such pathways .

Research Applications

This protein serves as a model for studying:

Bacterial Regulatory Mechanisms

  • Gene Expression: Investigating ramA operon dynamics, including interactions with RamR (a repressor) and RamA (an activator) .

  • Efflux Pump Regulation: Exploring potential roles in modulating acrAB or related pump systems.

Recombinant Protein Production

  • Host Optimization: Testing Corynebacterium glutamicum or other hosts for enhanced yield, as seen in adaptive evolution studies .

  • Protein Engineering: Assessing solubility and stability to improve purification efficiency .

Production Hurdles

  • Low Solubility: E. coli expression may yield insoluble aggregates, requiring refolding or chaperone co-expression .

  • Codon Bias: Rare codons in the sequence could limit translation efficiency; codon-optimized constructs may be necessary .

Functional Ambiguity

  • Lack of Biochemical Data: No published studies describe enzymatic activity, binding partners, or structural data.

  • Dependency on Hypothesis-Driven Research: Studies must rely on homology to ramA-associated genes for functional inference .

Future Directions

  • Structural Characterization: X-ray crystallography or NMR to elucidate 3D structure.

  • Functional Screening: Yeast two-hybrid or mass spectrometry to identify interaction partners.

  • Host Engineering: Leveraging adaptive evolution (e.g., C. glutamicum JH41) to improve recombinant yields .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: All proteins are shipped with standard 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 consolidate 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% and serves as a guideline.
Shelf Life
Shelf life depends on 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 manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Uncharacterized protein in ramA 3'region
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-82
Protein Length
full length protein
Species
Klebsiella pneumoniae
Target Protein Sequence
MKHPLETLLSAAGILLLALLSCLLLPAPSLGLTLAQKLVETFHMMDLNQLYTVLFCLWFL ALGAIEYLVLRWVWRRWFSLER
Uniprot No.

Target Background

Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What expression systems are most effective for producing this recombinant protein?

E. coli expression systems, particularly those utilizing the T7 promoter present in pET vectors, have proven effective for the production of this recombinant protein . The T7 promoter system is extremely popular for recombinant protein expression and can yield target protein representing up to 50% of the total cell protein in successful cases . The protein has been successfully expressed as a His-tagged construct in E. coli systems, facilitating downstream purification processes .

Alternative expression systems worth exploring include Chinese hamster ovary (CHO) cells, which are widely used for recombinant protein production in the pharmaceutical industry . The expression yield in mammalian systems can be significantly influenced by the choice of 3'untranslated regions (3'UTRs), with the human albumin 3'UTR demonstrating a 2-3 fold increase in expression levels for some proteins .

How can 3'UTR selection enhance expression levels of this uncharacterized protein?

The 3'untranslated region (3'UTR) selection can significantly impact recombinant protein expression levels. Research has demonstrated that replacing the endogenous 3'UTR with human albumin 3'UTR can increase expression 2-3 fold and mRNA levels up to 10-fold in mammalian expression systems . The albumin 3'UTR contains an AU-rich complex stem loop region in nucleotides 1-50 that appears critical for enhancing expression, as deletion of this region causes significant reductions in both protein expression and mRNA levels .

In contrast, replacement with immunoglobulin or chymotrypsinogen 3'UTR decreased expression levels in tested systems . This differential effect suggests that 3'UTR elements can be strategically selected to optimize production of recombinant proteins. When expressing the uncharacterized protein in ramA 3'region, researchers should consider testing different 3'UTR elements to determine which provides optimal expression levels for their specific experimental goals.

What purification strategies yield the highest purity and recovery of the functional protein?

For the recombinant uncharacterized protein in ramA 3'region expressed with a His-tag, immobilized metal affinity chromatography (IMAC) serves as the primary purification strategy . Based on experience with similar proteins, a multi-stage purification approach is recommended:

  • Initial capture using Ni-NTA or cobalt-based IMAC with optimized imidazole gradient elution

  • Intermediate purification using ion exchange chromatography based on the protein's theoretical pI

  • Polishing step using size exclusion chromatography to separate monomeric protein from aggregates

Buffer optimization is critical, particularly considering the protein's potential membrane association based on its sequence characteristics . For storage, a Tris-based buffer with 50% glycerol has been successfully used , though researchers should validate storage conditions for their specific application.

What computational approaches can predict functional characteristics of this uncharacterized protein?

In silico characterization represents a powerful first step in determining potential functions of uncharacterized proteins. The following methodological workflow is recommended:

  • Domain and motif analysis using tools like Pfam, SMART, and CDD to identify conserved functional elements

  • Structural prediction using AlphaFold2 or I-TASSER to generate 3D models for function inference

  • Sequence homology comparison with characterized proteins across bacterial species

  • Identification of conserved superfamilies, such as those identified in other uncharacterized proteins (e.g., HDC_protein, M34_PPEP, PBECR3, and SPASM superfamilies)

The analysis of conserved domains and superfamilies has proven valuable in predicting functional roles of uncharacterized proteins based on their conserved domains and motifs . For example, proteins containing the M34_PPEP domain may function as extracellular metalloproteases that could influence bacterial phenotypes, while SPASM domains often appear in peptide-modifying enzymes .

What experimental approaches best elucidate the function of uncharacterized proteins in bacterial systems?

A comprehensive experimental approach to characterize the function of this protein should include:

  • Gene knockout studies using CRISPR-Cas9 or homologous recombination to observe phenotypic effects

  • Transcriptomic and proteomic analysis comparing wild-type and knockout strains under various conditions

  • Protein-protein interaction studies using pull-down assays, bacterial two-hybrid systems, or co-immunoprecipitation

  • Subcellular localization studies using fluorescent protein fusions or immunofluorescence microscopy

For antimicrobial resistance evaluation, methodologies from studies of similar uncharacterized proteins suggest using AMRFinderPlus and ResFinder to identify potential resistance genes and virulence factors . These tools have successfully identified genes like vanZ1 (glycopeptide resistance) and blaCDD (beta-lactam resistance) in other uncharacterized protein studies .

How can researchers determine if this protein contributes to antimicrobial resistance pathways?

To determine potential contributions to antimicrobial resistance (AMR), researchers should employ a multi-faceted approach:

  • Comparative minimum inhibitory concentration (MIC) assays between wild-type and protein knockout strains against various antibiotics

  • Gene expression analysis of known AMR genes in the presence and absence of the uncharacterized protein

  • Proteomic interaction studies with known AMR regulators, particularly with the RamA protein

  • Computational AMR gene analysis using specialized tools like AMRFinderPlus, which can identify glycopeptide resistance proteins (vanZ1) and beta-lactamase genes (blaCDD)

Research on uncharacterized proteins in similar bacterial species has demonstrated that these proteins can play roles in virulence and antimicrobial resistance, making them potential targets for drug discovery or vaccine development .

What regulatory guidelines apply to research involving this recombinant protein?

Research involving the recombinant uncharacterized protein in ramA 3'region falls under the NIH Guidelines for Research Involving Recombinant or Synthetic Nucleic Acid Molecules . Specifically:

  • Expression in E. coli typically falls under Section III-E or III-D depending on the containment level required

  • If the protein is expressed in mammalian cells, different sections may apply based on the expression system

  • For experiments involving viable recombinant microorganisms tested on animals, Section III-D-4-a applies

Institutional Biosafety Committee (IBC) approval is generally required for recombinant DNA work, and researchers should consult their local IBC for specific institutional requirements . Additionally, research involving K. pneumoniae requires Biosafety Level 2 (BSL-2) containment practices due to its status as a potential pathogen.

What criteria should be considered when evaluating this protein as a potential vaccine or drug target?

When evaluating the uncharacterized protein in ramA 3'region as a potential therapeutic target, researchers should assess:

  • Non-homology to human proteins to avoid cross-reactivity (ideally <30% sequence identity)

  • Antigenicity and non-allergenicity profiles

  • Presence of virulence factors or involvement in pathogenicity

  • Accessibility of the protein to therapeutic agents (surface exposure)

  • Conservation across K. pneumoniae strains for broad-spectrum targeting

Studies of uncharacterized proteins in pathogenic bacteria have shown that approximately 36-41% may be antigenic and most are non-homologous to human proteins, making them potential vaccine candidates . The ability to identify virulence factors through computational analysis further enhances target selection capabilities.

How can protein stability and solubility be optimized during expression and purification?

Optimizing stability and solubility of the uncharacterized protein in ramA 3'region requires systematic troubleshooting:

  • Expression temperature optimization (typically 16-30°C), with lower temperatures often favoring proper folding

  • Induction conditions testing (IPTG concentration from 0.1-1.0 mM)

  • Co-expression with chaperones or T7 lysozyme to improve folding

  • Fusion tags evaluation (MBP, SUMO, or GST) if His-tag constructs show poor solubility

  • Buffer screening using thermal shift assays to identify stabilizing conditions

For storage, the protein has been successfully maintained in Tris-based buffer with 50% glycerol . Researchers should avoid repeated freeze-thaw cycles by storing working aliquots at 4°C for up to one week and longer-term storage at -20°C or -80°C .

What are the recommended approaches for studying protein-protein interactions involving this uncharacterized protein?

To identify and characterize protein-protein interactions involving the uncharacterized protein in ramA 3'region:

  • Affinity purification-mass spectrometry (AP-MS) using the His-tagged protein as bait

  • Bacterial two-hybrid or three-hybrid assays to screen for interaction partners

  • Surface plasmon resonance (SPR) or bio-layer interferometry (BLI) for kinetic analysis of identified interactions

  • Cross-linking mass spectrometry to identify interaction interfaces

  • Computational prediction of interaction partners through genomic context analysis and co-expression data

These methods can help identify if the protein interacts with antimicrobial resistance regulators like RamA or other proteins involved in virulence pathways. The information on interacting proteins is currently limited in existing databases , making this an important area for investigation.

How should researchers interpret conflicting results from different characterization methods?

When faced with conflicting results from different characterization methods, researchers should:

  • Evaluate methodological limitations of each approach (sensitivity, specificity, false positive/negative rates)

  • Consider biological context and physiological relevance of each experimental condition

  • Perform validation using orthogonal techniques that rely on different principles

  • Consult studies of related uncharacterized proteins where similar conflicts were resolved

For example, studies of uncharacterized proteins in C. difficile found that AMR analysis from NCBI and RAST yielded different results, with AMRFinderPlus identifying more AMR genes in whole genome sequences than in the predicted protein sequences alone . This illustrates the importance of using multiple complementary approaches when characterizing novel proteins.

What bioinformatic pipelines are most effective for comparative analysis across Klebsiella strains?

An effective bioinformatic pipeline for comparative analysis should include:

  • Whole genome sequence alignment using tools like Mauve or Progressive Mauve

  • Pangenome analysis using Roary or PGAP to identify core and accessory genes

  • SNP calling with tools like Snippy or GATK

  • Phylogenetic analysis using RAxML or IQ-TREE to establish evolutionary relationships

  • Comparative protein structure prediction using AlphaFold2 across strains

This approach can help determine the conservation of the uncharacterized protein in ramA 3'region across different K. pneumoniae strains and identify strain-specific variations that might affect function or antigenicity.

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.