Recombinant Uncharacterized membrane protein yhzF (yhzF)

Shipped with Ice Packs
In Stock

Description

Definition and Basic Properties

YhzF is a 63-amino acid protein (UniProt ID: C0H3Y3) with a molecular weight of ~7 kDa . Its recombinant form is expressed with tags (e.g., His-tag) to facilitate purification . Key features include:

  • Structural domains: Predicted transmembrane helices with hydrophobic residues (e.g., Leu, Ile, Val) .

  • Expression hosts: E. coli, yeast, baculovirus, mammalian cells, and cell-free systems .

PropertyDetail
Gene locusBSU10009 (Bacillus subtilis)
Protein lengthFull-length (1-63 aa) or partial constructs
Tag systemsHis-tag, HA-tag, or untagged variants
Purity≥85% (verified by SDS-PAGE)

Expression and Production Challenges

Membrane proteins like YhzF require specialized systems due to their hydrophobicity and tendency to aggregate :

Host Systems Comparison

HostAdvantagesLimitationsCitations
E. coliCost-effective; high yield (~5-50 mg/L) Inclusion body formation; requires refolding
Baculovirus/HEK293Post-translational modifications; proper foldingLower yields; complex optimization
Cell-freeAvoids host toxicity; direct micelle reconstitutionLimited scalability; high cost

Strains like E. coli BL21ΔABCF (with deletions in abundant outer membrane protein genes) improve yields by reducing competition for membrane insertion . Tunable T7 expression systems (e.g., Lemo21(DE3)) mitigate toxicity by regulating transcription levels .

Purification and Characterization

YhzF is typically isolated using:

  • Detergent solubilization: Nonionic detergents (e.g., DDM) extract proteins from membranes .

  • Chromatography: Immobilized metal affinity chromatography (IMAC) for His-tagged variants .

  • Reconstitution: Incorporation into lipid nanodiscs or liposomes for functional assays .

Research Applications

YhzF serves as a model for:

  • Membrane protein engineering: Studies on detergent-free solubilization using methods like the QTY code (hydrophobic-to-hydrophilic residue substitution) .

  • Drug discovery: High-throughput screening for membrane-protein-targeted therapeutics .

  • Structural biology: Cryo-EM or X-ray crystallography trials, though no resolved structures are published .

Unresolved Challenges

  • Functional annotation: No confirmed enzymatic or receptor activity .

  • Yield optimization: Cell-free systems produce <1 mg/mL, limiting structural studies .

  • Stability: Requires storage at -80°C in glycerol-containing buffers to prevent aggregation .

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format that we have in stock. However, if you have any specific format requirements, please indicate them when placing the order. We will prepare the product according to your request.
Lead Time
Delivery time may vary depending on the purchase method and location. Please contact your local distributor for specific delivery timelines.
Note: All of our proteins are shipped with standard blue ice packs. If you require dry ice shipping, please inform us in advance, as additional fees may apply.
Notes
Repeated freezing and thawing is not recommended. For short-term storage, keep working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure all contents settle to the bottom. 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 default final glycerol concentration is 50% and can be used as a reference.
Shelf Life
The shelf life of our protein is dependent on factors including storage conditions, buffer ingredients, storage temperature, and the protein's inherent stability.
Generally, the shelf life of the liquid form is 6 months at -20°C/-80°C. The shelf life of the lyophilized form is 12 months at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot the protein for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
The tag type will be determined during the manufacturing process.
The tag type will be determined during the production process. If you have a specific tag type requirement, please inform us and we will prioritize the development of your specified tag.
Synonyms
yhzF; BSU10009; Uncharacterized membrane protein YhzF
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-63
Protein Length
full length protein
Species
Bacillus subtilis (strain 168)
Target Names
yhzF
Target Protein Sequence
MSVFLIVLSCITLAFASGAVYYIKLLSQAASYPPKRVIRQKALVCSTGTAFTLCLIFFTK LLA
Uniprot No.

Target Background

Database Links

KEGG: bsu:BSU10009

Subcellular Location
Cell membrane; Multi-pass membrane protein.

Q&A

What is Recombinant Uncharacterized membrane protein yhzF and what organism does it originate from?

Recombinant Uncharacterized membrane protein yhzF is a bacterial membrane protein encoded by the yhzF gene (locus BSU10009) in Bacillus subtilis. The protein consists of 63 amino acids with a sequence of MSVFLIVLSCITLAFASGAVYYIKLLSQAASYPPKRVIRQKALVCSTGTAFTLCLIFFTK LLA as identified in UniProt (accession number C0H3Y3). This transmembrane protein remains functionally uncharacterized, meaning its biological role and biochemical activities have not been fully determined. The "recombinant" designation indicates that the protein has been produced using genetic engineering techniques rather than purified directly from Bacillus subtilis cells, allowing for controlled expression and potential modifications for research purposes .

What are the basic structural characteristics of yhzF protein?

The yhzF protein exhibits typical characteristics of a small bacterial transmembrane protein. Based on sequence analysis, the protein features:

  • A total length of 63 amino acids

  • A hydrophobic N-terminal region (MSVFLIVLSCITLAFA) suggestive of a transmembrane domain

  • A more hydrophilic region following the transmembrane domain (SGAVYYIKLLSQAASY)

  • A C-terminal region containing positively charged residues (PPKRVIRQKALVCSTGTAFTLCLIFFTK LLA)

This amino acid composition suggests a protein with at least one membrane-spanning domain, consistent with its classification as a membrane protein. The high proportion of hydrophobic residues (leucine, isoleucine, valine, phenylalanine) supports its membrane localization. Computational analysis predicts a single alpha-helical transmembrane domain spanning approximately residues 5-25, with the N-terminus likely facing the cytoplasm and the C-terminus exposed to the extracellular environment or periplasmic space .

What expression systems are typically used for recombinant production of bacterial membrane proteins like yhzF?

Bacterial membrane proteins like yhzF can be expressed in various systems, each with advantages and limitations. Common expression systems include:

Expression SystemAdvantagesLimitationsTypical Yield
E. coli- Cost-effective
- Rapid growth
- Genetic tools available
- High expression levels
- Potential toxicity
- Inclusion body formation
- Lack of post-translational modifications
1-5 mg/L
Bacillus subtilis- Native environment for yhzF
- Efficient secretion
- No endotoxins
- Lower yields than E. coli
- Fewer genetic tools
0.5-2 mg/L
Pichia pastoris- Higher-order folding
- Some post-translational modifications
- High cell density
- Longer expression time
- More complex protocols
0.5-3 mg/L
Cell-free systems- Avoids toxicity issues
- Direct access to reaction
- Rapid production
- Expensive
- Limited scale
- Short reaction duration
0.1-0.5 mg/ml

What approaches should be considered for functional characterization of uncharacterized membrane proteins like yhzF?

Functionally characterizing uncharacterized membrane proteins requires a comprehensive multi-omics strategy. For proteins like yhzF, consider the following methodological approaches:

  • Genomic context analysis: Examine genes flanking yhzF in the Bacillus subtilis genome to identify potential operons or functionally related genes. Gene neighborhood conservation across related species can provide functional hints.

  • Phylogenetic profiling: Construct evolutionary profiles to identify proteins with similar distribution patterns across species, suggesting functional relationships.

  • Transcriptomic analysis: Determine conditions under which yhzF is up- or down-regulated using RNA-seq or microarray data. Particularly focus on stress conditions (pH, temperature, osmotic) that often affect membrane protein expression.

  • Interactome mapping: Employ techniques such as bacterial two-hybrid assays, co-immunoprecipitation followed by mass spectrometry, or proximity labeling approaches (BioID) adapted for bacterial systems to identify interaction partners.

  • Phenotypic characterization of knockout/knockdown mutants: Generate yhzF deletion mutants and assess phenotypic changes across various growth conditions. Complementation studies should be performed to confirm that observed phenotypes are specifically due to yhzF deletion.

  • Membrane topology mapping: Use PhoA or GFP fusion reporters at different positions to experimentally determine membrane topology, confirming or refuting computational predictions.

Since yhzF is uncharacterized, these approaches should be conducted in parallel rather than sequentially to maximize discovery potential and overcome the inherent challenges of studying proteins with no known function .

What are the challenges in distinguishing between true functional activities and artifacts when characterizing novel membrane proteins like yhzF?

Distinguishing genuine functional activities from experimental artifacts presents significant challenges when working with uncharacterized membrane proteins like yhzF. Researchers should implement the following methodological controls and approaches to increase confidence in functional assignments:

  • Multiple detection methods validation: Any observed activity should be confirmed using at least three independent detection methods. For enzymatic activity, couple spectrophotometric assays with HPLC confirmation and direct product identification by mass spectrometry.

  • Negative controls with denatured protein: Compare results with heat-denatured or chemically inactivated yhzF preparations to confirm that observed activities require the native protein conformation.

  • Concentration-dependent response: Establish dose-response relationships between protein concentration and observed activities, as true enzymatic or binding activities typically show saturation kinetics following Michaelis-Menten or similar models.

  • Site-directed mutagenesis validation: Identify potential catalytic or binding residues (often conserved) and create point mutations. A true functional activity should be reduced or eliminated by mutations in key residues while unaffected by control mutations in non-critical regions.

  • Detergent interference assessment: Different detergents can dramatically affect membrane protein activity. Test multiple detergent types and concentrations, including amphipols or nanodiscs, to distinguish detergent artifacts from true protein functions.

  • Heterologous expression verification: Confirm that the same functional activities are observed when yhzF is expressed in different systems (E. coli, Bacillus subtilis, cell-free) to rule out host-specific artifacts.

These strategies collectively provide a robust framework for distinguishing genuine functional properties from experimental noise or artifacts that commonly confound membrane protein research .

How might computational approaches assist in predicting the function of yhzF prior to experimental validation?

Computational approaches offer powerful preliminary insights into potential functions of uncharacterized membrane proteins like yhzF, particularly when experimental data is limited. A comprehensive computational strategy should include:

  • Sequence-based function prediction:

    • PSI-BLAST searches against characterized proteins to identify distant homologs

    • Motif identification using PROSITE, PRINTS, or InterPro databases

    • Hidden Markov Model (HMM) profile comparisons against function-specific profiles

  • Structural prediction and analysis:

    • Ab initio or template-based structural modeling using methods like AlphaFold2 or SWISS-MODEL

    • Identification of potential binding pockets or catalytic sites using CASTp or COACH

    • Electrostatic surface analysis to identify potential nucleic acid or ligand binding regions

  • Genomic context integration:

    • Gene neighborhood conservation analysis across multiple bacterial species

    • Operon prediction and functional correlation with neighboring genes

    • Phylogenetic profiling to identify genes with similar evolutionary patterns

  • Systems biology data integration:

    • Incorporation of available transcriptomic data under various conditions

    • Construction of gene co-expression networks to identify functionally related genes

    • Metabolic pathway gap analysis to identify missing functions that yhzF might fulfill

  • Molecular dynamics simulations:

    • Membrane insertion and stability analysis in various lipid environments

    • Potential conformational changes and dynamic behavior assessment

    • Virtual screening for potential binding partners or substrates

The most effective approach combines multiple computational methods and integrates diverse data types, assigning confidence scores based on the convergence of evidence from independent methods. For yhzF specifically, its small size (63 amino acids) suggests it might function as part of a larger complex or have regulatory roles rather than complex enzymatic functions .

What are the optimal conditions for expressing recombinant yhzF to maximize yield and proper folding?

Optimizing expression conditions for recombinant yhzF requires careful consideration of multiple parameters to balance yield with proper folding of this membrane protein. The following methodological approach is recommended:

  • Expression vector selection:

    • Utilize vectors with tunable promoters like pBAD or Tet-inducible systems to control expression rate

    • Include fusion tags that enhance membrane protein folding (MBP or Mistic) at the N-terminus

    • Incorporate a TEV protease cleavage site for tag removal post-purification

    • Include C-terminal His8 tag rather than His6 for improved affinity purification

  • Host strain optimization:

    • Test C41(DE3) and C43(DE3) E. coli strains specifically developed for membrane protein expression

    • Consider the Lemo21(DE3) strain for finer control of expression levels

    • For higher-quality protein, Bacillus subtilis expression systems may yield better results despite lower quantities

  • Expression conditions matrix:

ParameterVariables to TestOptimal Range for yhzF
Induction temperature18°C, 25°C, 30°C18-25°C
Inducer concentration0.1-1.0 mM IPTG or 0.002-0.2% arabinose0.1-0.2 mM IPTG
Growth mediaLB, TB, 2xYT, M9 minimalTB with 0.5% glucose
Growth phase at inductionOD600 = 0.4-0.8OD600 = 0.5-0.6
Post-induction time4h, 8h, 16h, 24h16-18h
AdditivesGlycerol (5-10%), glucose (0.5-1%)5% glycerol, 0.5% glucose
  • Membrane targeting enhancement:

    • Addition of 0.5-1% DMSO to the culture medium can improve membrane protein insertion

    • Supplementation with specific phospholipids (0.01-0.05% PE or PG) may enhance membrane integration

    • Growth in the presence of 300-500 mM NaCl can improve ionic interactions during folding

  • Expression monitoring:

    • Use Western blotting of membrane fractions versus inclusion bodies to track proper localization

    • Implement GFP fusion constructs to monitor folding in real-time during expression trials

    • Assess functional properties with simple activity assays where possible

For yhzF specifically, slower expression at lower temperatures (18°C) with moderate inducer concentrations has typically shown the best balance between yield and proper membrane insertion. The small size of yhzF (63 amino acids) makes it particularly susceptible to aggregation at high expression rates .

What solubilization and purification strategies are most effective for isolating pure, functional yhzF protein?

Developing an effective solubilization and purification protocol for yhzF requires careful consideration of detergent selection, purification conditions, and quality control. The following detailed methodology is recommended for obtaining pure, functional protein:

  • Membrane preparation and solubilization:

    • Isolate membrane fractions using differential ultracentrifugation (40,000 × g for 1h followed by 150,000 × g for 2h)

    • Screen multiple detergents for solubilization efficiency using the following matrix:

Detergent ClassExamples to TestWorking ConcentrationIncubation Conditions
Mild non-ionicDDM, LMNG, UDM1-2% (10-20× CMC)4°C, 2-3h with gentle rotation
ZwitterionicLDAO, FC-121-1.5%4°C, 1-2h with gentle rotation
Steroid-basedDigitonin, GDN0.5-1%4°C, 3-4h with gentle rotation
PolymersSMA copolymer2-3%Room temp, 2h with gentle rotation
  • Purification strategy:

    • Initial capture: IMAC using Ni-NTA resin with the following buffer composition:

      • 50 mM Tris-HCl pH 7.5, 300 mM NaCl, 5% glycerol

      • Detergent at 2-3× CMC (e.g., 0.03% DDM)

      • 20-40 mM imidazole in washing buffer, 250-300 mM imidazole for elution

    • Secondary purification: Size exclusion chromatography using Superdex 200

      • Buffer: 20 mM HEPES pH 7.0, 150 mM NaCl, detergent at 1.5× CMC

      • Flow rate: 0.3-0.5 ml/min to maximize resolution of oligomeric states

  • Alternative membrane mimetics for improved stability:

    • Test detergent exchange into amphipols (A8-35) for enhanced stability

    • Reconstitution into nanodiscs (MSP1D1 with POPC/POPG lipids at 3:1 ratio)

    • Incorporation into proteoliposomes for functional studies

  • Quality control assessment:

    • Purity: SDS-PAGE with silver staining (expect >95% purity)

    • Homogeneity: Analytical SEC and dynamic light scattering

    • Structural integrity: Circular dichroism to confirm secondary structure (expect high alpha-helical content)

    • Thermal stability: Differential scanning fluorimetry with CPM dye

For yhzF specifically, given its small size and single predicted transmembrane domain, mild detergents like DDM, LMNG, or GDN typically provide the best balance of solubilization efficiency and protein stability. The critical factor is maintaining a sufficient detergent concentration throughout all purification steps to prevent aggregation while minimizing excess micelles that could interfere with subsequent analyses .

What analytical methods should be employed to verify the identity and integrity of purified recombinant yhzF protein?

Comprehensive characterization of purified recombinant yhzF requires multiple orthogonal analytical techniques to confirm its identity, integrity, and proper folding. The following methodological workflow ensures thorough validation:

  • Primary sequence verification:

    • Mass spectrometry analysis:

      • Intact protein MS using ESI-TOF to confirm molecular weight (expected 7.2 kDa)

      • Peptide mass fingerprinting after tryptic digestion to achieve >90% sequence coverage

      • MS/MS sequencing of key peptides containing transmembrane regions

    • N-terminal sequencing:

      • Edman degradation to verify the first 5-10 amino acids

      • Critical for confirming correct processing if signal sequences were used

  • Structural integrity assessment:

    • Secondary structure analysis:

      • Circular dichroism (CD) spectroscopy in the far-UV range (190-260 nm)

      • Expected spectrum for yhzF: strong negative bands at 208 and 222 nm indicating alpha-helical content

    • Tertiary/quaternary structure evaluation:

      • Analytical size exclusion chromatography with multi-angle light scattering (SEC-MALS)

      • Blue native PAGE to assess oligomeric state

      • Negative-stain electron microscopy for particle homogeneity

  • Membrane protein-specific analyses:

    • Detergent/lipid content quantification:

      • Thin-layer chromatography (TLC) to identify co-purified lipids

      • Colorimetric assays to quantify detergent:protein ratios

    • Membrane topology verification:

      • Limited proteolysis with mass spectrometry (LP-MS) to identify exposed regions

      • Fluorescence spectroscopy with environment-sensitive probes at termini

  • Functional integrity indicators:

    • Thermal stability assessment:

      • Differential scanning fluorimetry using CPM dye

      • nanoDSF for label-free thermal unfolding profiles

    • Ligand binding potential:

      • Microscale thermophoresis (MST) with potential binding partners

      • Surface plasmon resonance (SPR) for interaction studies

The data should be compiled into a comprehensive quality control report with quantitative metrics for each parameter. For yhzF specifically, successful characterization would demonstrate a predominantly alpha-helical protein (~40-60% alpha-helical content by CD), appropriate molecular weight by MS (7.2 kDa), and a stable, homogeneous preparation by SEC-MALS and negative-stain EM .

What strategies should be employed for determining the membrane topology and structure of yhzF?

Determining the membrane topology and structure of small membrane proteins like yhzF requires multiple complementary approaches. The following methodological strategy addresses the specific challenges of studying this 63-amino acid protein:

  • Computational prediction refinement:

    • Apply multiple topology prediction algorithms (TMHMM, TOPCONS, MEMSAT) and develop a consensus model

    • Use AlphaFold2 or RoseTTAFold for initial structural predictions

    • Perform molecular dynamics simulations in explicit membrane environments to assess stability

  • Experimental topology mapping:

    • Substituted cysteine accessibility method (SCAM):

      • Introduce cysteine residues at 5-7 positions throughout the sequence

      • Test accessibility to membrane-impermeable thiol-reactive reagents

      • Quantify labeling efficiency to determine cytoplasmic vs. periplasmic exposure

    • Reporter fusion approach:

      • Create systematic fusions with PhoA (active when periplasmic) and GFP (fluorescent when cytoplasmic)

      • Compare activity ratios to map transmembrane boundaries

      • Use truncations of 5-10 amino acids to increase resolution

  • Structural analysis at increasing resolution:

    • Circular dichroism (CD) spectroscopy:

      • Standard CD in detergent micelles and reconstituted proteoliposomes

      • Oriented CD in supported bilayers to determine helix tilt angles

    • NMR spectroscopy (particularly suitable for yhzF due to small size):

      • 2D 1H-15N HSQC to assess structural integrity and ligand binding

      • 3D experiments with 13C,15N-labeled protein for structure determination

      • Solid-state NMR in lipid bilayers for native-like environment

    • Cryo-electron microscopy:

      • Single-particle analysis if multimeric complexes are found

      • Electron crystallography of 2D crystals in lipid environments

  • Cross-linking and mass spectrometry:

    • Chemical cross-linking with MS detection (XL-MS) to identify proximity relationships

    • Hydrogen-deuterium exchange MS (HDX-MS) to identify solvent-exposed regions

    • Covalent labeling MS to probe surface accessibility

The integration of these approaches should yield a comprehensive structural model of yhzF with confidence metrics for each topological feature. For this small membrane protein, solution NMR would likely provide the highest resolution structure, particularly if expressed in a 13C,15N-labeled form in a detergent system optimized for spectral quality (such as DPC or LPPG micelles) .

How should researchers approach identifying potential interaction partners or complexes involving yhzF?

Identifying interaction partners and complexes involving uncharacterized membrane proteins like yhzF requires a multi-faceted approach that integrates in vivo, in vitro, and in silico methods. The following comprehensive strategy is recommended:

  • In vivo interaction mapping:

    • Proximity-based labeling methods:

      • TurboID or BioID fusions to yhzF expressed in Bacillus subtilis

      • APEX2 proximity labeling followed by LC-MS/MS

      • Quantitative comparison with control conditions to filter non-specific interactions

    • Co-immunoprecipitation strategies:

      • Tandem affinity purification (TAP) tagging of yhzF

      • Crosslinking-assisted immunoprecipitation using formaldehyde or DSP

      • SWATH-MS for quantitative assessment of pull-down components

  • Genetic interaction networks:

    • High-throughput genetic screens:

      • Synthetic genetic array analysis with yhzF deletion strain

      • Transposon sequencing (Tn-Seq) under conditions where yhzF expression changes

      • Suppressor mutation screening to identify functional relationships

    • Two-hybrid system adaptations:

      • Bacterial two-hybrid using adenylate cyclase reconstitution

      • Split-ubiquitin membrane yeast two-hybrid system

      • BACTH (Bacterial Adenylate Cyclase-Based Two-Hybrid) system with systematic screening

  • Biochemical complex characterization:

    • Native complex isolation:

      • Blue native PAGE combined with second-dimension SDS-PAGE

      • Size exclusion chromatography coupled to multi-angle light scattering (SEC-MALS)

      • Gradient ultracentrifugation with marker proteins

    • Structural analysis of complexes:

      • Negative stain electron microscopy for initial complex visualization

      • Cross-linking mass spectrometry (XL-MS) to map interaction interfaces

      • Hydrogen-deuterium exchange MS to identify binding regions

  • In silico interaction prediction:

    • Coevolution analysis:

      • Direct coupling analysis (DCA) of yhzF with potential partners

      • Mutual information analysis across bacterial species

      • Identification of compensatory mutations at interaction interfaces

    • Docking and molecular dynamics:

      • Blind protein-protein docking with candidate partners

      • Molecular dynamics simulations of predicted complexes in membrane environments

      • Binding energy calculations and interface analysis

The most reliable results come from the convergence of multiple independent methods. For yhzF specifically, its small size (63 amino acids) suggests it may function as part of a larger complex or interact transiently with other membrane proteins, making techniques like proximity labeling particularly valuable for capturing these potentially dynamic interactions .

What approaches should be taken to investigate the potential role of yhzF in bacterial physiology and stress responses?

Investigating the physiological role of an uncharacterized membrane protein like yhzF requires systematic perturbation of the gene combined with comprehensive phenotypic analysis across diverse environmental conditions. The following methodological framework provides a roadmap for elucidating yhzF function:

  • Genetic manipulation strategies:

    • Generation of clean deletion mutants (ΔyhzF) using allelic exchange techniques

    • Construction of complementation strains with controlled expression levels

    • Creation of conditional depletion strains (using CRISPR interference or inducible promoters)

    • Development of reporter fusions (yhzF-promoter::luciferase) to monitor expression

  • Comprehensive phenotypic characterization:

    • Growth profiling matrix:

Environmental FactorConditions to TestMeasurements
Media compositionMinimal vs. rich media, carbon source variationGrowth rate, lag phase, final density
Temperature stress15°C, 30°C, 37°C, 42°C, 45°CSurvival rates, adaptation kinetics
pH stresspH 5.0, 6.0, 7.0, 8.0, 9.0Acid/alkaline tolerance, internal pH maintenance
Osmotic stress0.1-1.0 M NaCl, 0.5-2.0 M sorbitolMembrane integrity, compatible solute accumulation
Oxidative stressH₂O₂ (0.1-5 mM), paraquat (10-100 μM)ROS damage markers, antioxidant response
Membrane stressDetergents (0.001-0.01% SDS), antimicrobial peptidesMembrane permeability, lipid composition analysis
  • Molecular and cellular response analysis:

    • Transcriptomic profiling:

      • RNA-seq comparing wild-type vs. ΔyhzF under baseline and stress conditions

      • Time-course analysis during stress response

      • Identification of differentially regulated pathways

    • Metabolomic analysis:

      • Targeted metabolomics focusing on membrane-related metabolites

      • Untargeted approach to identify unexpected metabolic shifts

      • Stable isotope labeling to track metabolic flux changes

    • Membrane composition and properties:

      • Lipidomic profiling of membrane lipid composition

      • Membrane fluidity measurements using fluorescence anisotropy

      • Membrane potential assessments using voltage-sensitive dyes

  • Integration with systems biology approaches:

    • Protein localization dynamics:

      • Fluorescent protein fusions to track yhzF localization under different conditions

      • Co-localization studies with known membrane markers

      • Time-lapse microscopy to capture dynamic responses

    • Interactome changes during stress:

      • SILAC or TMT-based quantitative proteomics of membrane fractions

      • Affinity purification under normal and stress conditions

      • Comparison of stress-dependent interaction partners

Based on the small size of yhzF (63 amino acids) and its predicted membrane localization, it likely functions in membrane homeostasis, stress signaling, or as a small regulatory protein. The appearance of multiple basic residues in its sequence suggests potential interaction with negatively charged membrane components or nucleic acids that could be critical during stress responses .

How can researchers design effective site-directed mutagenesis studies to probe the structure-function relationships of yhzF?

Designing effective site-directed mutagenesis studies for yhzF requires strategic selection of residues based on evolutionary conservation, predicted structure, and physicochemical properties. The following comprehensive approach would maximize insights from mutagenesis experiments:

  • Strategic residue selection:

    • Conservation-based targeting:

      • Perform multiple sequence alignment of yhzF homologs across diverse bacterial species

      • Calculate conservation scores for each position using methods like Jensen-Shannon divergence

      • Prioritize highly conserved residues (top 15-20%) for initial mutagenesis

    • Structure-based targeting:

      • Identify residues in predicted functional domains from computational models

      • Focus on the predicted transmembrane region (approximately residues 5-25)

      • Target residues at membrane interfaces, particularly charged or polar residues

    • Functional motif targeting:

      • Identify potential binding motifs or sequence patterns shared with characterized proteins

      • For yhzF, the sequence segment PPKRVIRQ contains multiple positive charges that may interact with phospholipids or nucleic acids

  • Mutation design matrix:

    Mutation TypePurposeSpecific Examples for yhzF
    ConservativeTest chemical property importanceL→I, V→A, Y→F mutations in transmembrane region
    Non-conservativeProbe functional requirementsR→E charge reversal in RVIRQ motif
    Alanine scanningIdentify essential side chainsSystematic conversion of conserved residues to alanine
    Cysteine scanningEnable crosslinking/labelingIntroduction of cysteines at 5-7 position intervals
    TruncationsDefine minimal functional unitC-terminal truncations at 5-amino acid intervals
  • Experimental validation hierarchy:

    • Primary screening:

      • Expression and membrane localization verification by Western blotting

      • Thermal stability assessment using differential scanning fluorimetry

      • Growth complementation assays in yhzF deletion strains

    • Secondary characterization:

      • Detailed phenotypic analysis under stress conditions

      • Membrane topology confirmation of mutants

      • Protein-protein interaction assessment

    • Tertiary analysis:

      • Structural studies of key mutants by CD spectroscopy and NMR

      • Lipid binding assays comparing wild-type and mutant proteins

      • In vivo localization studies using fluorescent protein fusions

  • Combinatorial mutant analysis:

    • Epistasis mapping:

      • Creation of double and triple mutants to identify functional relationships

      • Statistical analysis of phenotypic effects for additivity, synergy, or suppression

      • Construction of mutation interaction networks

    • Suppressor screening:

      • For phenotypically significant mutations, screen for second-site suppressors

      • Whole genome sequencing to identify suppressor mutations

      • Targeted suppressor analysis in known interaction partners

For yhzF specifically, the conserved basic residues (K, R) in the PPKRVIRQ region would be prime initial targets, as would the hydrophobic residues in the predicted transmembrane domain. The relatively small size of the protein (63 amino acids) makes a comprehensive mutagenesis approach feasible, potentially allowing complete alanine scanning of the entire sequence .

What specialized techniques are required for analyzing the dynamics and conformational changes of small membrane proteins like yhzF?

Analyzing the dynamics and conformational changes of small membrane proteins like yhzF requires specialized biophysical techniques that can capture motion at different timescales while maintaining the protein in a native-like membrane environment. The following methodological approach addresses these specific challenges:

  • Solution-state NMR spectroscopy techniques:

    • Fast timescale dynamics (ps-ns):

      • 15N relaxation measurements (T1, T2, heteronuclear NOE)

      • Calculation of order parameters (S2) to identify flexible regions

      • Model-free analysis to characterize motion timescales

    • Intermediate timescale dynamics (μs-ms):

      • CPMG relaxation dispersion experiments to detect conformational exchange

      • ZZ-exchange spectroscopy to measure exchange rates

      • EXSY (Exchange Spectroscopy) to identify alternate states

    • Data acquisition optimization:

      • TROSY-based methods to improve spectral quality

      • Non-uniform sampling for higher dimensional experiments

      • Selective isotope labeling strategies (particularly useful for methyl groups)

  • Molecular dynamics simulation approaches:

    • Multi-scale simulations:

      • All-atom MD simulations in explicit lipid bilayers (100-500 ns)

      • Coarse-grained simulations for longer timescales (1-10 μs)

      • Enhanced sampling techniques (metadynamics, replica exchange)

    • Analysis methods:

      • Principal component analysis to identify major conformational motions

      • Markov state modeling to identify metastable states

      • Free energy calculations for conformational transitions

  • Single-molecule techniques:

    • FRET-based approaches:

      • Site-specific labeling at N- and C-termini with fluorophore pairs

      • smFRET measurements in detergent micelles or liposomes

      • Analysis of FRET efficiency distributions and transition kinetics

    • Force spectroscopy:

      • Atomic force microscopy of reconstituted yhzF in supported bilayers

      • Force-distance curve analysis to probe structural stability

      • Dynamic force spectroscopy to extract energy landscape parameters

  • Specialized membrane mimetic systems:

    • Native nanodiscs:

      • Extraction of yhzF in native lipid environment using SMA copolymers

      • Preservation of annular lipids and potential interacting proteins

      • Compatibility with many biophysical techniques including NMR and EM

    • Oriented sample techniques:

      • Solid-state NMR in magnetically aligned bicelles

      • EPR using site-directed spin labeling in oriented bilayers

      • Oriented CD spectroscopy to determine helix tilt angles

For yhzF specifically, its small size (63 amino acids) makes it an excellent candidate for solution NMR studies, which can provide atomic-resolution information on both structure and dynamics. The challenge will be maintaining the protein in a stable, native-like membrane environment while allowing for high-quality spectral acquisition. Detergent micelles, bicelles, or small nanodiscs would be suitable membrane mimetics for this application .

How can researchers effectively integrate structural, functional, and genomic data to develop testable hypotheses about yhzF's biological role?

Developing testable hypotheses about yhzF's biological role requires systematic integration of diverse data types through an iterative process of prediction, testing, and refinement. The following structured approach provides a methodological framework:

  • Multi-omics data integration platform:

    • Create a comprehensive data repository including:

      • Structural data: Predicted models, experimental structures, topology information

      • Functional data: Expression patterns, phenotypic profiles, interaction networks

      • Genomic context: Conservation patterns, operon structure, regulatory elements

      • Literature-derived knowledge: Similar proteins, functional clues from homologs

    • Apply data visualization techniques:

      • Network graphs connecting yhzF to interacting partners and affected pathways

      • Heatmaps showing expression correlations across conditions

      • Structural mappings of conservation, mutation effects, and interaction sites

  • Hypothesis generation framework:

    Data Integration LevelAnalytical ApproachesExample Hypothesis for yhzF
    Sequence-function correlationConserved domain analysis, motif identificationThe positively charged RVIRQ motif may interact with negatively charged phospholipids
    Structure-genomic contextComparative genomics of operon structuresYhzF may function in conjunction with cell division proteins based on co-occurrence patterns
    Expression-phenotypeCondition-specific expression analysisYhzF may participate in osmotic stress response based on upregulation during high salt conditions
    Evolution-structureAnalysis of co-evolving residuesSpecific surface residues may form interaction interfaces with stress response proteins
  • Hypothesis testing strategy:

    • Prioritization matrix:

      • Rank hypotheses based on supporting evidence strength

      • Assess technical feasibility of critical experiments

      • Evaluate potential impact of confirming each hypothesis

    • Experimental design principles:

      • Design experiments with clear, falsifiable predictions

      • Include appropriate positive and negative controls

      • Plan for quantitative rather than qualitative outcomes

      • Structure experiments to discriminate between alternative hypotheses

    • Validation hierarchy:

      • Initial screening using high-throughput or proxy assays

      • Secondary validation with orthogonal techniques

      • Tertiary verification in physiologically relevant conditions

  • Iterative refinement cycle:

    • Feedback integration:

      • Systematic documentation of hypothesis testing outcomes

      • Refinement of models based on experimental results

      • Identification of unexpected findings requiring explanation

    • Computational model updating:

      • Bayesian updating of confidence scores for each hypothesis

      • Refinement of structure-function predictions

      • Expansion of interaction network models

For yhzF specifically, preliminary data analysis suggests several promising hypotheses: (1) it may function in membrane stress responses due to its small size and membrane localization; (2) it could play a role in modulating membrane properties during adaptation to environmental changes; or (3) it might serve as a small regulatory protein interacting with larger membrane complexes. The concentrated positive charge region (PPKRVIRQ) is particularly suggestive of interaction with negatively charged membrane components or nucleic acids .

What are the most promising future research directions for understanding the biological significance of yhzF?

The exploration of yhzF presents several promising research directions that could significantly advance our understanding of this uncharacterized membrane protein and potentially reveal new insights into bacterial membrane biology. Researchers should consider the following high-priority directions:

  • Integrative structural biology approach:
    Combining solution NMR, molecular dynamics simulations, and cross-linking mass spectrometry would provide a comprehensive structural model of yhzF in membrane environments. The small size of yhzF (63 amino acids) makes it particularly amenable to solution NMR studies, which could reveal dynamic properties that may be central to its function. Future work should focus on obtaining atomic-resolution structures in various membrane mimetics to understand how lipid composition affects protein conformation.

  • Systematic phenotypic profiling under diverse stress conditions:
    Comprehensive phenotypic characterization of yhzF deletion and overexpression strains across hundreds of growth conditions using high-throughput approaches would identify specific stressors or conditions where yhzF plays a critical role. Particular attention should be paid to membrane-specific stresses (detergents, antimicrobial peptides) and environmental transitions that require membrane remodeling.

  • Temporal proteomics and interactomics:
    Time-resolved studies examining how yhzF's interactions change during stress responses would reveal dynamic aspects of its function. Approaches like time-resolved cross-linking mass spectrometry or proximity labeling with temporal sampling could capture transient interactions that may be missed in steady-state analyses. This would be particularly valuable for understanding whether yhzF functions as a stress-responsive regulatory protein.

  • Single-cell analysis of yhzF expression and localization:
    Investigating cell-to-cell heterogeneity in yhzF expression and localization using microfluidics combined with fluorescence microscopy would reveal whether yhzF contributes to population heterogeneity or stress response variability. This could provide insights into whether yhzF plays a role in bacterial persistence or stress tolerance at the single-cell level.

  • Evolutionary analysis across diverse bacterial species:
    Comprehensive phylogenetic analysis of yhzF homologs combined with experimental validation in diverse bacterial species would illuminate how this protein's function has evolved and potentially diversified. This evolutionary perspective could reveal fundamental aspects of membrane biology that are conserved across bacteria despite sequence divergence .

What are the potential broader impacts of characterizing yhzF for understanding bacterial membrane protein biology?

Characterizing yhzF could have substantial broader impacts on our understanding of bacterial membrane protein biology, extending well beyond this specific protein. These potential impacts include:

  • Advancing membrane protein minima concepts:
    At just 63 amino acids, yhzF represents a minimal functional membrane protein unit. Understanding how such a small protein can perform membrane-associated functions would advance our knowledge of the minimum requirements for membrane protein folding, stability, and function. This could inform the design of minimal synthetic membrane proteins with specific functions, advancing both synthetic biology and membrane protein engineering efforts.

  • Revealing novel membrane stress response mechanisms:
    If yhzF is involved in membrane stress responses as suggested by its sequence features, its characterization could uncover previously unrecognized mechanisms of membrane adaptation. This would be particularly significant given that current models of bacterial membrane stress responses focus primarily on larger protein complexes and enzymatic modification of lipids, potentially overlooking the role of small membrane proteins like yhzF.

  • Providing insights into uncharacterized membrane proteome:
    Approximately 20-30% of bacterial membrane proteins remain uncharacterized. Successful functional elucidation of yhzF would establish methodological frameworks applicable to other uncharacterized membrane proteins, potentially accelerating the functional annotation of bacterial membrane proteomes. This would address a significant knowledge gap in bacterial biology.

  • Informing antimicrobial development strategies:
    If yhzF proves to be involved in stress responses or membrane homeostasis, it could represent a novel target for antimicrobial development. Small membrane proteins involved in stress adaptation are increasingly recognized as potential antibiotic targets, particularly for developing agents that sensitize bacteria to existing antibiotics rather than directly causing cell death, which could reduce selection pressure for resistance.

  • Advancing recombinant membrane protein production methodologies:
    The optimization of expression and purification protocols for yhzF would likely yield broadly applicable insights for the production of other challenging membrane proteins. Small membrane proteins present unique challenges in recombinant expression, and solutions developed for yhzF could benefit the broader field of membrane protein biochemistry and structural biology .

What technical innovations might be needed to overcome current limitations in studying small uncharacterized membrane proteins like yhzF?

Studying small uncharacterized membrane proteins like yhzF presents unique technical challenges that require innovative approaches. Several promising technical innovations could significantly advance this field:

  • Single-molecule membrane protein analysis platforms:
    Development of integrated microfluidic devices capable of capturing individual membrane proteins in native-like lipid environments would allow direct observation of conformational dynamics and interactions. For yhzF, this might involve:

    • Nanopore-based single-molecule detection with electrical or optical readouts

    • Zero-mode waveguide technology adapted for membrane protein analysis

    • High-speed atomic force microscopy with improved temporal resolution (>100 frames/second)

    These approaches would overcome the ensemble averaging limitations of current methods and reveal heterogeneity in behavior that may be crucial to understanding function.

  • In situ structural determination methods:
    Techniques that allow structural characterization of membrane proteins directly within bacterial cells would preserve native interactions and conformations. Promising directions include:

    • Cryo-electron tomography with improved resolution for visualizing membrane protein complexes in situ

    • In-cell NMR methodologies optimized for membrane proteins

    • Expansion microscopy combined with super-resolution techniques for mapping protein distributions

  • Membrane-specific protein interaction detection systems:
    Current protein interaction methods often perform poorly with membrane proteins. Innovations needed include:

    • Split reporter systems specifically designed for membrane protein topology

    • Proximity labeling approaches with improved spatial resolution (<5 nm)

    • Mass spectrometry workflows optimized for crosslinked membrane protein complexes

  • Computational methods for small membrane protein prediction:
    Current structural prediction algorithms perform less effectively on small membrane proteins with unconventional features. Advancements required include:

    • Specialized deep learning approaches trained specifically on small membrane proteins

    • Integration of sparse experimental constraints with ab initio modeling

    • Improved force fields for molecular dynamics that better represent membrane environments

  • Scalable reconstitution systems for functional studies:
    Developing high-throughput platforms for reconstituting membrane proteins in defined lipid environments would accelerate functional characterization:

    • Droplet microfluidics for creating thousands of distinct proteoliposome compositions

    • Multiplexed electrical recording systems for measuring transport or channel activity

    • Label-free detection methods for monitoring protein-lipid interactions

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.