Recombinant Haemophilus influenzae Heme exporter protein D (ccmD)

Shipped with Ice Packs
In Stock

Description

Molecular Characterization

Gene and Protein Details

  • Gene name: ccmD (Cytochrome c maturation gene D)

  • Protein length: 69 amino acids (UniProt ID: P0ABM6)

  • Sequence:
    MTPAFASWNEFFAMGGYAFFVWLAVVMTVIPLVVLVVHSVMQHRAILRGVAQQRAREARLRAAQQQEAA

Key Findings:

  • CcmD is dispensable for heme transfer to CcmE but essential for releasing holo-CcmE from the transporter complex .

  • Deletion of the central hydrophobic domain abolishes function, while the C-terminal domain requires a net positive charge for activity .

Research Applications

  • Cytochrome c biogenesis studies: Used to dissect heme export mechanisms in Gram-negative bacteria .

  • Membrane protein interaction assays: Employed in co-immunoprecipitation and topology mapping experiments .

Comparative Analysis with Homologs

OrganismGene NameFunctionKey Difference
Rhodobacter capsulatushelDHeme exportShares 36% sequence identity with ccmD
Escherichia coliccmD/yojMABC transporter holo-CcmE release factorContains additional domains (e.g., ECK2190)

Challenges and Future Directions

  • Unresolved questions: Structural dynamics of the CcmABCD complex and precise role of the GXY motif remain under investigation .

  • Therapeutic potential: While not directly targeted, insights into ccmD could inform antimicrobial strategies targeting heme trafficking .

Product Specs

Form
Lyophilized powder
Note: We will prioritize shipping the format currently in stock. However, if you have a specific format requirement, please indicate it during order placement. We will prepare the product accordingly.
Lead Time
Delivery time may vary depending on the purchase method and location. Please consult your local distributors for specific delivery timelines.
Note: All protein shipments are standardly packaged with blue ice packs. If you require dry ice packaging, please inform us in advance, as additional charges will apply.
Notes
Repeated freezing and thawing is not recommended. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend briefly centrifuging the vial before opening to ensure the contents settle at the bottom. Reconstitute the protein in deionized sterile 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%, which can serve as a reference.
Shelf Life
Shelf life is influenced by various factors, including storage conditions, buffer composition, temperature, and the protein's inherent stability.
Generally, liquid formulations have a shelf life of 6 months at -20°C/-80°C. Lyophilized formulations have a shelf life of 12 months at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type will be determined during the manufacturing process.
Tag type is determined during production. If you have a specific tag type preference, please inform us, and we will prioritize developing the specified tag.
Synonyms
ccmD; HI_1092; Heme exporter protein D; Cytochrome c-type biogenesis protein CcmD
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-67
Protein Length
full length protein
Species
Haemophilus influenzae (strain ATCC 51907 / DSM 11121 / KW20 / Rd)
Target Names
ccmD
Target Protein Sequence
MFFQTWSDFFNMGGYGFYVWLSYAVSLVAVIALIVQSVKQRKTVLQNVLREKQREERLQQ ANKGNTL
Uniprot No.

Target Background

Function
Essential for heme export to the periplasm, a crucial step in the biogenesis of c-type cytochromes.
Database Links

KEGG: hin:HI1092

STRING: 71421.HI1092

Protein Families
CcmD/CycX/HelD family
Subcellular Location
Cell inner membrane; Single-pass membrane protein.

Q&A

What is Heme exporter protein D (ccmD) and what is its function in Haemophilus influenzae?

Heme exporter protein D (ccmD) is a critical component of the cytochrome c-type biogenesis system in Haemophilus influenzae. This small membrane protein (69 amino acids) functions within the cytochrome c maturation (CCM) pathway, facilitating the export and delivery of heme to apocytochromes. The protein is essential for proper heme trafficking and the assembly of functional c-type cytochromes, which are crucial for the respiratory chain and energy metabolism in H. influenzae .

As a heme auxotroph, H. influenzae cannot synthesize heme but has evolved sophisticated mechanisms for acquiring, transporting, and utilizing exogenous heme, with ccmD playing a specialized role in this process. The ccmD protein operates as part of a larger heme handling mechanism that enables the bacterium to survive in heme-limited environments of the human respiratory tract .

How is recombinant ccmD typically expressed and purified?

Recombinant Haemophilus influenzae ccmD is commonly expressed in several heterologous systems with the following methodological approaches:

Expression systems:

  • E. coli (most common)

  • Yeast expression systems

  • Baculovirus-infected insect cells

  • Mammalian cell expression systems

Typical expression protocol:

  • The ccmD gene is PCR-amplified from H. influenzae genomic DNA

  • The gene is cloned into an expression vector with an N-terminal or C-terminal affinity tag (commonly His-tag)

  • Expression is typically driven by an inducible promoter system (e.g., T7 promoter with IPTG induction)

  • The recombinant protein is expressed with high yield after optimization of induction conditions

Purification approach:

  • Bacterial cells are harvested and lysed (often requiring detergents due to membrane association)

  • The recombinant protein is purified by affinity chromatography (commonly Ni-NTA for His-tagged constructs)

  • Further purification may include size exclusion chromatography or ion exchange chromatography

  • Final purity is typically ≥85% as determined by SDS-PAGE analysis

The purified protein is often stored in Tris/PBS-based buffer with 6% trehalose at pH 8.0 to maintain stability .

What experimental approaches are recommended for studying ccmD protein-protein interactions?

When investigating the protein-protein interactions of Haemophilus influenzae ccmD, researchers should consider implementing a multi-method approach for comprehensive characterization. The following experimental design is recommended:

Primary methods:

  • Co-immunoprecipitation (Co-IP)

    • Express ccmD with an epitope tag (His, FLAG, etc.)

    • Lyse cells under conditions that preserve protein-protein interactions

    • Pull down ccmD using tag-specific antibodies

    • Identify interacting partners by mass spectrometry

  • Bacterial two-hybrid system

    • Particularly useful for membrane-associated proteins like ccmD

    • Construct fusion proteins with split reporter domains

    • Screen for interactions based on functional complementation

    • Validate with targeted pairwise testing of suspected interaction partners

  • Crosslinking coupled with mass spectrometry

    • Treat intact bacterial cells with membrane-permeable crosslinkers

    • Isolate ccmD complexes by affinity purification

    • Analyze by LC-MS/MS to identify crosslinked peptides

    • Map interaction interfaces based on crosslinked residues

Complementary approaches:

  • Surface Plasmon Resonance (SPR)

    • Immobilize purified ccmD on a sensor chip

    • Flow potential interaction partners over the surface

    • Measure real-time binding kinetics and affinities

  • Microscale Thermophoresis (MST)

    • Label purified ccmD with a fluorescent tag

    • Titrate with unlabeled potential binding partners

    • Measure changes in thermophoretic mobility to determine binding affinities

Experimental design considerations:

  • Single-subject experimental design with careful controls is appropriate when initial screening identifies potential interactions

  • Include negative controls (non-related proteins) and positive controls (known interacting partners from the Ccm system)

  • Validate interactions through reciprocal pull-downs with tagged versions of identified partners

  • Consider the membrane environment when designing experiments, as detergent choice can significantly impact results

This multi-method approach helps overcome limitations of individual techniques and provides robust evidence for protein-protein interactions involving the ccmD protein.

How should researchers design single-subject experiments to study ccmD function in heme export?

When investigating ccmD function in heme export using single-subject experimental design, researchers should implement the following methodological framework:

Experimental design structure:

  • Baseline phase (A):

    • Measure heme transport/cytochrome c maturation in wild-type H. influenzae

    • Establish stable baseline measurements across multiple time points

    • Quantify cytochrome c levels using spectroscopic methods

    • Measure heme content using appropriate biochemical assays

  • Intervention phase (B):

    • Introduce genetic modification (ccmD knockout or controlled expression)

    • Continue measurements using identical protocols as baseline

    • Maintain all other variables constant

    • Collect sufficient data points to establish new stable pattern

  • Return to baseline or second intervention (A' or C):

    • Either complementation with wild-type ccmD (return to baseline)

    • Or introduction of mutant ccmD variants (second intervention)

    • Continue measurements with identical protocols

Key methodological considerations:

  • Repeated measurements: Collect multiple data points in each phase to understand variability and determine treatment effects

  • Control for extraneous variables: Maintain consistent growth conditions, heme availability, and other environmental factors

  • Randomization: Where applicable, randomize the order of measurements to reduce bias

  • Visual analysis: Plot time-series data to visually inspect changes across phases

  • Statistical analysis: Apply appropriate single-subject statistics (e.g., percentage of non-overlapping data points, standardized mean difference)

Example experimental design table:

PhaseConditionDurationMeasurementsFrequency
AWild-type2 weeksCytochrome c levels, Heme content, Growth rateDaily
BccmD knockout2 weeksCytochrome c levels, Heme content, Growth rateDaily
CccmD complementation2 weeksCytochrome c levels, Heme content, Growth rateDaily
DPoint mutant ccmD2 weeksCytochrome c levels, Heme content, Growth rateDaily

This design allows researchers to establish causal relationships between ccmD variants and functional outcomes by using each experimental subject as its own control, which is particularly valuable when working with specialized bacterial strains .

What controls should be included when expressing recombinant H. influenzae ccmD?

When expressing recombinant Haemophilus influenzae ccmD, incorporating comprehensive controls is essential to ensure experimental validity and reliable interpretation of results. The following control framework should be implemented:

Expression system controls:

  • Negative expression control

    • Empty vector-transformed host cells processed identically

    • Verifies background signals and non-specific binding during purification

    • Essential for distinguishing true ccmD-specific effects from artifacts

  • Positive expression control

    • Well-characterized protein with similar size/properties to ccmD

    • Confirms expression system and purification workflow functionality

    • Typically a known protein from the Ccm system (e.g., ccmC or ccmE)

  • Induction controls

    • Uninduced cultures of the ccmD-expressing construct

    • Partially induced cultures (titration of inducer)

    • Validates induction system and helps optimize expression conditions

Protein quality controls:

  • Tag-only control

    • Expression of the affinity tag portion without ccmD

    • Distinguishes tag-mediated from ccmD-specific effects

    • Essential when studying protein-protein interactions

  • Known mutation controls

    • Expression of ccmD with characterized mutations

    • Inactive variant (negative functional control)

    • Hyperactive variant if available (positive functional control)

Methodological controls:

  • SDS-PAGE and Western blot controls

    • Molecular weight standards

    • Purified ccmD standard (if available)

    • Anti-tag antibody controls for specificity verification

  • Functional assay controls

    • Heat-denatured ccmD (negative control)

    • Establish dose-response relationships with varying ccmD concentrations

    • Substrate-free reactions

Recombinant integrity verification:

  • DNA sequence verification

    • Confirm complete ccmD sequence before expression

    • Verify absence of unintended mutations

  • Protein sequence verification

    • Mass spectrometry analysis of purified protein

    • Peptide mapping to confirm identity

  • Endotoxin and contaminant testing

    • Especially important for downstream functional studies

    • Limulus Amebocyte Lysate (LAL) assay for endotoxin quantification

This comprehensive control framework ensures that the expressed recombinant ccmD protein is authentic, properly folded, and suitable for downstream applications, while minimizing experimental artifacts .

How can researchers investigate the interaction between ccmD and other components of the cytochrome c maturation (Ccm) system?

Investigating the interactions between ccmD and other Ccm system components requires sophisticated approaches that account for the membrane-associated nature of these proteins. The following comprehensive methodology is recommended:

Genetic interaction analysis:

  • Synthetic genetic arrays

    • Generate a matrix of double mutants (ccmD with other Ccm components)

    • Quantify genetic interactions through growth phenotypes or cytochrome c levels

    • Identify synthetic lethal, synthetic sick, or suppressor relationships

  • Suppressor screens

    • Introduce random mutations in a ccmD mutant background

    • Select for restoration of cytochrome c maturation

    • Sequence suppressors to identify functional relationships

Biochemical interaction characterization:

  • Membrane-based pull-down assays

    • Express epitope-tagged ccmD in H. influenzae

    • Solubilize membranes with compatible detergents (e.g., DDM, LMNG)

    • Perform pull-downs and identify co-purifying proteins by mass spectrometry

    • Validate interactions with reciprocal pull-downs

  • In vitro reconstitution

    • Purify individual Ccm components including ccmD

    • Reconstitute into proteoliposomes or nanodiscs

    • Assess complex formation by size exclusion chromatography

    • Measure functional activities (heme transport, cytochrome c maturation)

Structural approaches:

  • Crosslinking mass spectrometry

    • Use membrane-permeable crosslinkers in intact cells

    • Purify ccmD complexes and analyze by MS

    • Map crosslinked residues to identify interaction interfaces

    • Build structural models of ccmD-Ccm component interactions

  • Cryo-electron microscopy

    • Purify intact Ccm complexes containing ccmD

    • Optimize sample preparation for single-particle cryo-EM

    • Determine structure of multi-protein complexes

    • Locate ccmD within the larger assembly

Functional validation:

  • Site-directed mutagenesis followed by functional assays

    • Mutate predicted interaction interfaces on ccmD

    • Express mutants and assess effects on:

      • Protein-protein interactions (pull-downs)

      • Heme transport (spectroscopic assays)

      • Cytochrome c maturation (enzymatic activity)

  • In vivo proximity labeling

    • Fuse ccmD to a proximity labeling enzyme (BioID, APEX)

    • Express in H. influenzae under native conditions

    • Identify labeled proteins that are in close proximity to ccmD

    • Compare with biochemical interaction data

This multi-faceted approach yields complementary data on the spatial, functional, and physical interactions between ccmD and other Ccm components, providing a comprehensive understanding of its role in the cytochrome c maturation system .

What approaches can be used to analyze potential contradictions in ccmD functional data across different studies?

Analyzing contradictions in ccmD functional data across different studies requires a systematic framework for identifying, categorizing, and resolving discrepancies. The following methodology helps researchers address such contradictions:

Systematic contradiction analysis framework:

  • Identify and categorize contradictions

    • Apply the (α, β, θ) notation system to classify contradictions :

      • α = number of interdependent items/variables

      • β = number of contradictory dependencies reported

      • θ = minimal number of Boolean rules needed to assess contradictions

    • Categorize contradictions as either:

      • Methodological (arising from different techniques)

      • Biological (reflecting true strain differences)

      • Interpretive (resulting from different analytical approaches)

  • Contradiction resolution methodology

    Data quality assessment:

    • Evaluate experimental controls used in each study

    • Assess statistical power and sample sizes

    • Examine reproducibility within each study

    • Consider laboratory-specific factors that might influence results

    Methodological comparison:

    • Create a comprehensive table mapping differences in:

      • Experimental conditions (temperature, media, growth phase)

      • Expression systems (E. coli strains, vector systems)

      • Purification methods (detergents, buffers, tags)

      • Functional assay conditions (substrates, detection methods)

    Biological context evaluation:

    • Analyze H. influenzae strain differences

    • Compare genomic context of ccmD

    • Assess potential post-translational modifications

    • Consider environmental adaptations of different isolates

Quantitative contradiction resolution:

  • Meta-analytical approach

    • Standardize outcomes across studies

    • Calculate effect sizes with confidence intervals

    • Perform heterogeneity analysis (I² statistic)

    • Use forest plots to visualize cross-study variations

  • Boolean minimization

    • Define Boolean equations representing contradictory results

    • Apply Quine-McCluskey algorithm to minimize contradictions

    • Identify minimal set of rules that explain observed variations

Practical resolution strategies:

  • Direct experimental reconciliation

    • Design experiments that specifically address contradictions

    • Replicate critical experiments from conflicting studies under identical conditions

    • Test multiple H. influenzae strains side-by-side

    • Employ multiple complementary techniques to examine the same phenomenon

Example contradiction analysis table:

StudyStrainExpression SystemPurification MethodFunctional OutcomePossible Explanation for Contradiction
Study AH. influenzae RdE. coli BL21(DE3)Ni-NTA, native conditionsHigh heme binding affinityUse of full-length protein with native conformation
Study BH. influenzae NTHiE. coli C41(DE3)His-tag, denaturing conditionsLow heme binding affinityDenaturation affecting protein structure
Study CH. influenzae clinical isolateCell-free expressionFLAG-tag purificationModerate heme binding with strain-specific effectsClinical isolate genetic variation

This structured approach to analyzing contradictions helps researchers identify the most likely explanations for discrepancies and design targeted experiments to resolve them, ultimately leading to a more coherent understanding of ccmD function .

How does ccmD function compare across different Haemophilus species, and what experimental design best captures these differences?

Comparing ccmD function across different Haemophilus species requires a carefully designed experimental approach that accounts for phylogenetic relationships while maintaining methodological consistency. The following comprehensive research design is recommended:

Comparative genomic framework:

  • Phylogenetic analysis

    • Construct phylogenetic trees based on:

      • ccmD sequence alone

      • Whole-genome SNP analysis

      • Concatenated housekeeping genes

    • Map functional differences onto phylogenetic relationships

    • Identify species-specific adaptations versus conserved functions

  • Genomic context analysis

    • Compare organization of ccm gene clusters across species

    • Identify synteny or rearrangements

    • Analyze promoter regions and regulatory elements

    • Assess presence of complementary heme acquisition systems

Experimental comparative design:

  • Standardized expression and purification

    • Clone ccmD from multiple Haemophilus species:

      • H. influenzae (including typeable and non-typeable strains)

      • H. haemolyticus

      • H. parainfluenzae

      • H. ducreyi

    • Express all variants in the same expression system

    • Use identical purification protocols

    • Verify protein integrity by consistent analytical methods

  • Functional characterization matrix

    • Test all ccmD variants using standardized assays:

      • Heme binding affinity (surface plasmon resonance)

      • Cytochrome c maturation efficiency

      • Interaction with other Ccm components

      • Membrane integration properties

    • Maintain identical experimental conditions across all species

Cross-species complementation:

  • Gene replacement studies

    • Generate ccmD knockout strains in multiple Haemophilus species

    • Complement with ccmD genes from different species

    • Quantify restoration of function using standardized assays

    • Identify species-specific functional elements

Specialized experimental design elements:

  • Competition assays in heme-limited conditions

    • Co-culture different Haemophilus species

    • Limit heme availability to create selection pressure

    • Measure relative fitness using species-specific markers

    • Determine if ccmD variants confer competitive advantages

  • Heme scavenging capacity analysis

    • Similar to the hemophilin studies in H. haemolyticus

    • Compare heme acquisition efficiency across species

    • Analyze if ccmD contributes to interspecies competition for heme

    • Test if ccmD variants show different affinities under various heme concentrations

Comprehensive data integration:

  • Multi-parameter comparison table

SpeciesccmD Sequence Identity (%)Heme Binding Affinity (Kd)Cross-species Complementation Efficiency (%)Cytochrome c Production (relative units)Ecological Niche
H. influenzae Rd100 (reference)x.x × 10⁻⁷ M100 (self)100 (reference)Respiratory tract
H. influenzae NTHiXXx.x × 10⁻⁷ MXXXXRespiratory tract
H. haemolyticusXXx.x × 10⁻⁷ MXXXXRespiratory tract
H. parainfluenzaeXXx.x × 10⁻⁷ MXXXXOral cavity
H. ducreyiXXx.x × 10⁻⁷ MXXXXGenital mucosa

This comprehensive experimental design enables researchers to distinguish species-specific adaptations from conserved functions, correlate ccmD sequence variations with functional differences, and understand how these differences contribute to the ecological niches occupied by different Haemophilus species .

What are the recommended methods for optimizing recombinant ccmD expression and purification?

Optimizing recombinant Haemophilus influenzae ccmD expression and purification requires systematic troubleshooting and refinement of multiple parameters. The following methodological workflow provides a comprehensive approach:

Expression optimization strategy:

  • Vector and construct design

    • Test multiple affinity tags (His, GST, MBP) at both N and C termini

    • Evaluate different promoter strengths (T7, tac, araBAD)

    • Optimize codon usage for expression host

    • Consider fusion partners to enhance solubility

  • Expression host selection

    • Compare standard E. coli strains (BL21, Rosetta, C41/C43)

    • Test expression in cell-free systems

    • Evaluate specialized hosts for membrane proteins

    • Screen multiple clones for expression level variability

  • Culture condition optimization

    • Systematic testing of:

      • Induction temperature (15°C, 25°C, 30°C, 37°C)

      • Inducer concentration (IPTG: 0.1-1.0 mM range)

      • Media composition (LB, TB, autoinduction media)

      • Cell density at induction (OD₆₀₀: 0.4-1.0)

      • Post-induction duration (2-24 hours)

Membrane protein extraction optimization:

  • Lysis buffer screening

    • Test multiple buffer systems (Tris, HEPES, phosphate)

    • Optimize pH range (7.0-8.5)

    • Evaluate salt concentrations (100-500 mM NaCl)

    • Include stabilizing additives (glycerol 5-20%, trehalose 5-10%)

  • Detergent selection

    • Screen detergent panel:

      • Mild detergents (DDM, LMNG, DMNG)

      • Harsh detergents (SDS, sarkosyl)

      • Zwitterionic detergents (LDAO, FC-12)

    • Optimize detergent concentration (1-5× CMC)

    • Test detergent mixtures when necessary

Purification protocol optimization:

  • Initial capture optimization

    • For His-tagged constructs:

      • Compare Ni-NTA, TALON, HisTrap columns

      • Optimize imidazole concentrations in wash and elution buffers

      • Evaluate linear vs. step gradients for elution

    • For other tags, optimize corresponding affinity matrices

  • Secondary purification

    • Size exclusion chromatography:

      • Select appropriate column matrix (Superdex 75/200)

      • Optimize flow rate and sample loading

    • Ion exchange chromatography when applicable

    • Evaluate detergent exchange during purification

Protein quality assessment:

  • Analytical quality control

    • Purity assessment: SDS-PAGE, silver staining

    • Identity confirmation: Western blot, mass spectrometry

    • Homogeneity analysis: DLS, analytical SEC

    • Stability testing: thermal shift assays, time-course activity

Optimization results table:

ParameterTested RangeOptimal ConditionEffect on YieldEffect on Purity
Expression temperature15-37°C25°C3-fold increase vs. 37°CMinimal effect
IPTG concentration0.1-1.0 mM0.5 mMPlateau above 0.5 mMHigher purity at lower IPTG
Induction time2-24 hours16 hoursLinear increase up to 16hDecreased after 20h
DetergentDDM, LMNG, FC-12DDM (1%)LMNG (80% of DDM yield)LMNG (higher purity)
Buffer pH7.0-8.58.020% drop below pH 7.5Minimal effect
NaCl concentration100-500 mM300 mMDecreased at extremesHigher purity at 300 mM

This systematic optimization approach typically results in purified recombinant ccmD with ≥85% purity as determined by SDS-PAGE, suitable for structural and functional studies .

What analytical techniques are most effective for assessing ccmD-heme interactions?

Characterizing ccmD-heme interactions requires a multi-technique approach to fully understand binding parameters, structural changes, and functional implications. The following analytical methods provide complementary data on these interactions:

Spectroscopic methods:

  • UV-visible absorption spectroscopy

    • Primary method for detecting heme binding

    • Monitor Soret band (400-420 nm) and Q-bands (500-600 nm)

    • Quantify binding through titration experiments

    • Determine binding stoichiometry and approximate affinity

    • Distinguish between ferric (Fe³⁺) and ferrous (Fe²⁺) heme states

  • Circular dichroism (CD) spectroscopy

    • Far-UV CD (190-250 nm): monitor protein secondary structure changes upon heme binding

    • Near-UV CD (250-320 nm): detect tertiary structure alterations

    • Visible CD (350-650 nm): characterize the heme environment in the complex

  • Resonance Raman spectroscopy

    • Directly probe heme coordination state

    • Identify axial ligands (histidine coordination is typical in H. influenzae)

    • Distinguish between 5- and 6-coordinate heme

    • Detect strain in the porphyrin ring structure

Binding thermodynamics and kinetics:

  • Isothermal titration calorimetry (ITC)

    • Gold standard for determining binding thermodynamics

    • Provides binding affinity (Kd), stoichiometry (n), enthalpy (ΔH), and entropy (ΔS)

    • No labeling required

    • Requires significant amounts of purified protein

  • Surface plasmon resonance (SPR)

    • Measures real-time binding kinetics (kon and koff)

    • Determines binding affinity

    • Requires immobilization of either ccmD or heme-conjugate

    • Can detect binding under various buffer conditions

  • Fluorescence spectroscopy

    • Monitor intrinsic tryptophan fluorescence quenching upon heme binding

    • Determine binding constants in solution

    • Perform stopped-flow measurements for fast binding kinetics

Structural characterization:

  • X-ray crystallography

    • Determine high-resolution structure of ccmD-heme complex

    • Identify precise heme coordination geometry

    • Reveal protein conformational changes upon binding

    • Similar to approach used for hemophilin

  • Nuclear magnetic resonance (NMR) spectroscopy

    • Map heme binding site through chemical shift perturbations

    • Study dynamics of the complex in solution

    • Investigate paramagnetic effects of heme iron

    • Determine solution structure of smaller ccmD constructs

Specialized techniques:

  • Electron paramagnetic resonance (EPR)

    • Characterize the electronic state of heme iron

    • Distinguish high-spin vs. low-spin configurations

    • Identify the coordination environment

    • Detect interactions with nearby amino acids

  • Mass spectrometry approaches

    • Hydrogen-deuterium exchange MS: map conformational changes

    • Cross-linking MS: identify residues in proximity to heme

    • Native MS: determine complex stoichiometry and stability

Data integration and validation:

To effectively characterize ccmD-heme interactions, researchers should integrate data from multiple techniques. Below is a representative data table showing how complementary techniques provide a comprehensive binding profile:

ParameterTechniqueResultComplementary TechniqueValidation Result
Binding affinity (Kd)ITCx.x × 10⁻⁷ MSPRx.x × 10⁻⁷ M
Binding stoichiometryITCn = XNative MSn = X
Coordination stateResonance Raman5-coordinate, His ligationX-ray crystallographyHis-XX as axial ligand
Heme iron oxidation stateUV-visFe³⁺ (ferric)EPRHigh-spin Fe³⁺
Binding-induced conformational changeFar-UV CDIncreased α-helical contentHDX-MSProtected regions in helices X-Y

This multi-technique approach provides robust characterization of ccmD-heme interactions, ensuring reliable data through cross-validation of key parameters .

What are the best approaches for developing a ccmD knockout system in Haemophilus influenzae?

Developing an effective ccmD knockout system in Haemophilus influenzae requires careful consideration of this organism's particular genetic characteristics and transformation efficiency. The following comprehensive methodology provides options tailored to different research goals:

Traditional knockout strategies:

  • Insertional inactivation

    • Design strategy:

      • PCR-amplify ccmD gene with flanking regions (~500 bp each side)

      • Clone into a suicide vector for H. influenzae

      • Insert antibiotic resistance cassette (kanamycin or spectinomycin) into ccmD coding region

      • Ensure cassette contains transcriptional terminators to prevent polar effects

    • Transformation and selection:

      • Use natural transformation competence of H. influenzae

      • Select transformants on appropriate antibiotic media

      • Verify insertion by PCR and sequencing

    • Similar to approach used for hemophilin gene knockout in H. haemolyticus

  • Allelic replacement (clean deletion)

    • Design strategy:

      • Construct deletion cassette with upstream and downstream homology regions

      • Include counter-selectable marker (e.g., sacB)

      • Perform two-step recombination process

    • Process:

      • First recombination introduces the entire construct

      • Second recombination removes vector backbone and wild-type gene

      • Select for loss of counter-selectable marker

    • Verification:

      • PCR across deletion junction

      • Whole-genome sequencing to confirm clean deletion

Advanced genetic engineering approaches:

  • CRISPR/Cas9-based gene editing

    • System components:

      • Cas9 expression construct optimized for H. influenzae

      • sgRNA targeting ccmD sequence

      • Repair template with homology arms

    • Delivery methods:

      • Electroporation of ribonucleoprotein complexes

      • Transient plasmid expression

    • Selection:

      • CRISPR-induced lethality drives high efficiency without selection

      • Optional: include temporary antibiotic marker flanked by FRT sites

  • Conditional knockout systems

    • Inducible repression:

      • Replace native ccmD promoter with tetracycline-responsive promoter

      • Repress expression by adding anhydrotetracycline

      • Monitor gradual depletion of ccmD

    • Protein destabilization:

      • Fuse ccmD to destabilization domain (DD)

      • Stabilize with Shield-1 ligand

      • Remove ligand to induce protein degradation

Control systems and validation:

  • Complementation systems

    • Construct design:

      • Wild-type ccmD under native or inducible promoter

      • Integrate at neutral site or maintain on stable plasmid

      • Include epitope tag for expression verification

    • Controls:

      • Empty vector control

      • Point mutant ccmD variants (non-functional)

      • ccmD from related Haemophilus species

  • Validation approaches

    • Genomic:

      • PCR verification

      • Whole-genome sequencing

      • RNA-seq to confirm knockout and assess polar effects

    • Proteomic:

      • Western blot (if antibodies available)

      • Targeted proteomics (SRM/MRM mass spectrometry)

    • Phenotypic:

      • Growth curves under aerobic vs. anaerobic conditions

      • Cytochrome c spectra and activity assays

      • Heme utilization efficiency

Knockout strain characterization table:

StrainGenetic ModificationGrowth Rate (% of WT)Cytochrome c Content (% of WT)Heme Utilization (% of WT)Complementation Rescue?
Wild-typeNone100100100N/A
ΔccmDccmD deletionXXXXXXYes
ΔccmD + ccmDComplemented deletionXXXXXXN/A
ΔccmD + ccmD(H23A)Point mutant complementationXXXXXXNo
ccmD-DDDestabilization domain fusion100 (-Shield-1) XX (+Shield-1)100 (-Shield-1) XX (+Shield-1)100 (-Shield-1) XX (+Shield-1)N/A

This comprehensive approach to developing ccmD knockout systems provides researchers with multiple strategies adaptable to different research questions, from basic functional studies to detailed mechanistic investigations .

How might ccmD function be leveraged in understanding bacterial competition for heme resources?

Heme exporter protein D (ccmD) plays a crucial role in cytochrome c maturation and therefore in bacterial energy metabolism. Understanding its function can provide insights into bacterial competition for limited heme resources, particularly in the context of host-pathogen and bacteria-bacteria interactions. The following research directions can leverage ccmD function in this context:

Bacterial competition frameworks:

  • Co-culture competition models

    • Design experimental systems similar to hemophilin studies :

      • Co-culture H. influenzae with ccmD variants alongside other respiratory pathogens

      • Restrict heme availability to create competition

      • Measure relative fitness and population dynamics

      • Determine if ccmD expression levels or variants confer competitive advantages

  • In vivo competition studies

    • Develop animal models of respiratory colonization:

      • Inoculate with mixtures of wild-type and ccmD-modified strains

      • Track colonization dynamics in heme-restricted environments

      • Measure competitive index in different anatomical niches

      • Correlate ccmD function with in vivo fitness

Molecular mechanism investigations:

  • Comparative ccmD efficacy studies

    • Compare heme utilization efficiency across species:

      • Measure growth rates under heme limitation

      • Quantify cytochrome c maturation per unit of available heme

      • Determine minimal heme requirements for various species

      • Identify species with superior heme utilization via ccmD function

  • Heme sequestration mechanisms

    • Investigate if ccmD system can be modified to enhance heme sequestration:

      • Engineer ccmD variants with higher binding affinity

      • Test if overexpression creates a heme sink effect

      • Compare with dedicated heme sequestration systems like hemophilin

      • Assess if engineered strains can outcompete wild-type bacteria

Applications for microbiome engineering:

  • Probiotic development approach

    • Similar to H. haemolyticus inhibiting NTHi :

      • Engineer commensal Haemophilus strains with enhanced ccmD function

      • Test ability to reduce colonization by pathogenic species

      • Evaluate safety and stability of modified strains

      • Develop delivery systems for respiratory tract application

  • Synthetic competition systems

    • Design artificial consortia with varying ccmD capabilities:

      • Create defined bacterial communities with different heme acquisition systems

      • Monitor population dynamics under controlled conditions

      • Model interaction networks and competition outcomes

      • Identify emergent properties in complex communities

Data integration framework:

  • Competition prediction model

    • Develop multivariate analysis of heme-dependent competition:

SpeciesccmD Expression LevelHeme Acquisition SystemsCytochrome DiversityGrowth Rate Under Heme LimitationCompetitive Fitness
H. influenzaeReference (1×)Multiple (hgp, hxu, hem)HighModerateModerate
H. haemolyticusVariable (0.5-3×)Limited + hemophilinModerateVariable by strainStrain-dependent
Engineered strain AHigh (5×)EnhancedHighFastHigh
Engineered strain BKnockout (0×)Alternative pathwaysReducedSlowLow

This research direction offers significant potential for understanding bacterial competition in host environments and developing novel approaches to manage pathogenic colonization through manipulation of heme acquisition and utilization systems .

What experimental design would best evaluate the potential of targeting ccmD for antimicrobial development?

Evaluating ccmD as a potential antimicrobial target requires a comprehensive experimental approach spanning target validation, drug discovery, and preclinical assessment. The following research design provides a structured framework for this investigation:

Target validation phase:

  • Essentiality determination

    • Conditional knockout systems:

      • Develop tetracycline-regulated ccmD expression

      • Determine viability upon ccmD depletion under various conditions

      • Quantify growth inhibition and cytochrome c maturation defects

    • Saturating transposon mutagenesis:

      • Perform Tn-seq analysis across multiple growth conditions

      • Identify conditions where ccmD disruption is lethal

      • Compare essentiality in laboratory vs. host-mimicking conditions

  • Chemical validation

    • Design dominant-negative ccmD variants:

      • Create point mutations at key functional residues

      • Express in wild-type background

      • Determine if they inhibit native ccmD function

    • Develop peptide inhibitors of ccmD interactions:

      • Identify interaction interfaces with other Ccm components

      • Design peptides mimicking these interfaces

      • Test ability to disrupt cytochrome c maturation

High-throughput screening design:

  • Assay development

    • Primary screening assays:

      • Cell-based reporter system (e.g., cytochrome c-dependent luciferase)

      • In vitro ccmD-binding assays (fluorescence polarization)

      • Thermal shift assays with purified ccmD

    • Secondary confirmation assays:

      • Growth inhibition in H. influenzae

      • Cytochrome c spectral analysis

      • Oxygen consumption measurements

  • Compound library screening

    • Design diverse screening campaigns:

      • Natural product libraries

      • Fragment-based approaches

      • Structure-based virtual screening

      • Repurposing FDA-approved drugs

Lead optimization framework:

  • Structure-activity relationship studies

    • Medicinal chemistry approach:

      • Synthesize analogs of hit compounds

      • Determine minimal pharmacophore

      • Optimize potency, selectivity, and drug-like properties

    • Structural biology support:

      • Co-crystallize ccmD with lead compounds

      • Perform NMR binding studies

      • Use computational modeling to guide optimization

  • Target selectivity assessment

    • Evaluate effects on human heme-binding proteins:

      • Test against panel of human hemoproteins

      • Assess mitochondrial cytochrome maturation

      • Determine toxicity in mammalian cell cultures

Preclinical evaluation design:

  • Efficacy studies

    • In vitro microbiological assessment:

      • Determine MIC against diverse H. influenzae strains

      • Assess bactericidal vs. bacteriostatic activity

      • Evaluate resistance development frequency

    • Ex vivo models:

      • Human respiratory epithelial cell co-culture

      • Artificial sputum medium assays

      • Biofilm inhibition studies

  • Pharmacological evaluation

    • ADME properties:

      • Design appropriate formulations for respiratory delivery

      • Measure lung tissue penetration

      • Determine plasma and tissue half-life

    • Toxicology assessment:

      • Conduct respiratory irritation studies

      • Evaluate effects on lung microbiome

      • Assess systemic toxicity

PhaseKey ExperimentsSuccess CriteriaGo/No-Go Decision Points
Target ValidationConditional knockout phenotyping>80% growth inhibition upon ccmD depletionNo growth phenotype = No-Go
Assay DevelopmentReporter system Z'-factorZ' > 0.5 in 384-well formatPoor assay performance = No-Go
Hit IdentificationPrimary screen of ≥100,000 compounds≥0.1% hit rate with ≥50% inhibition<10 confirmed hits = No-Go
Lead OptimizationSAR with ≥50 analogsCompounds with IC₅₀ <1 μMNo potency improvement = No-Go
SelectivityHuman hemoprotein panel>50-fold selectivity vs. human proteinsPoor selectivity = No-Go
EfficacyMIC against clinical isolatesMIC ≤4 μg/mL for >90% of strainsPoor activity spectrum = No-Go
SafetyRespiratory toxicity studiesNo significant epithelial damage at 10× MICToxicity at therapeutic levels = No-Go

This comprehensive experimental design provides a clear pathway for evaluating ccmD as an antimicrobial target, with defined criteria for progression and decision points throughout the discovery and development process .

How can researchers validate that recombinant ccmD retains native-like structure and function?

Validating that recombinant ccmD maintains its native-like structure and function is crucial for ensuring experimental reliability. The following comprehensive validation framework provides multiple complementary approaches:

Structural validation:

  • Secondary structure analysis

    • Circular dichroism (CD) spectroscopy:

      • Compare far-UV CD spectra with predicted secondary structure

      • Analyze thermal stability profiles (melting temperatures)

      • Compare with other characterized Ccm proteins if available

    • Fourier-transform infrared spectroscopy (FTIR):

      • Complementary assessment of secondary structure elements

      • Particularly useful for membrane-associated proteins

  • Tertiary structure assessment

    • Intrinsic fluorescence spectroscopy:

      • Monitor tryptophan/tyrosine accessibility and environment

      • Compare with denatured controls to verify folded state

    • Limited proteolysis:

      • Observe proteolytic fragment patterns

      • Properly folded proteins show resistance to digestion

      • Compare patterns between recombinant and native protein (if available)

  • Quaternary structure evaluation

    • Size-exclusion chromatography:

      • Verify homogeneity and oligomeric state

      • Detect aggregation or improper assembly

    • Native PAGE or Blue Native PAGE:

      • Assess oligomerization without denaturing conditions

      • Compare migration pattern with expected molecular weight

Functional validation:

  • Heme binding characterization

    • UV-visible spectroscopy:

      • Monitor characteristic spectral changes upon heme binding

      • Determine binding stoichiometry and affinity

      • Compare with literature values if available

    • Resonance Raman spectroscopy:

      • Verify correct heme coordination state

      • Identify axial ligand identity

      • Compare with known heme-binding proteins

  • Protein-protein interaction assessment

    • Pull-down assays:

      • Test interactions with other Ccm components

      • Verify specific vs. non-specific binding

      • Compare with known interaction controls

    • Surface plasmon resonance:

      • Determine binding kinetics to interaction partners

      • Verify specificity through competition experiments

  • Functional complementation

    • Genetic complementation:

      • Express recombinant ccmD in ccmD knockout strain

      • Measure restoration of cytochrome c maturation

      • Compare with native ccmD expression

    • In vitro reconstitution:

      • Reconstitute Ccm system components including recombinant ccmD

      • Assess cytochrome c maturation activity

      • Compare with systems using native components

Native-like environment testing:

  • Membrane mimetic systems

    • Proteoliposome reconstitution:

      • Incorporate ccmD into artificial liposomes

      • Test functional activity in membrane environment

      • Compare various lipid compositions

    • Nanodiscs or lipid nanodiscs:

      • Provide defined membrane-like environment

      • Enable biophysical characterization in native-like context

  • Comparative analysis with native protein

    • Side-by-side comparison:

      • Extract native ccmD from H. influenzae (if feasible)

      • Compare biochemical and biophysical properties

      • Identify any differences for further optimization

Validation scoring matrix:

Validation MethodParameters MeasuredAcceptance CriteriaResult for Recombinant ccmDResult for Native ccmD (if available)
CD SpectroscopySecondary structure compositionConsistent with predictionα-helix: XX%, β-sheet: XX%α-helix: XX%, β-sheet: XX%
Thermal StabilityMelting temperature (Tm)Single transition, Tm >40°CTm = XX°CTm = XX°C
Heme BindingDissociation constant (Kd)Kd within 2-fold of literature valueKd = X.X × 10⁻⁷ MKd = X.X × 10⁻⁷ M
Proteolytic ResistanceFragment patternLimited number of stable fragmentsX major fragmentsX major fragments
Functional ComplementationCytochrome c levels>80% restoration of activityXX% of wild-type100%

This comprehensive validation framework enables researchers to systematically evaluate whether recombinant ccmD retains native-like structure and function, providing confidence in experimental results obtained with the recombinant protein .

What strategies can address data reproducibility challenges when studying Haemophilus influenzae ccmD?

Ensuring data reproducibility when studying Haemophilus influenzae ccmD requires systematic approaches to address variability sources across experimental workflows. The following comprehensive strategy framework provides practical solutions:

Experimental design reproducibility:

  • Standardized protocols development

    • Create detailed standard operating procedures (SOPs) covering:

      • Bacterial strain maintenance and growth conditions

      • Expression system preparation and induction

      • Protein purification with specific buffer compositions

      • Functional assays with step-by-step procedures

    • Include specific equipment settings and calibration methods

    • Document all reagent sources, lot numbers, and preparation dates

    • Implement electronic laboratory notebooks for complete record-keeping

  • Statistical power and experimental design

    • Incorporate appropriate experimental design principles:

      • A priori power analysis to determine required replicates

      • Randomization of sample processing order

      • Inclusion of appropriate positive and negative controls

      • Blinding of samples during analysis when feasible

    • Apply single-subject experimental design principles when appropriate

    • Document all exclusion criteria before experiments begin

Biological system reproducibility:

  • Strain and construct documentation

    • Maintain comprehensive strain information:

      • Complete genotypic and phenotypic characterization

      • Documented passage history and storage conditions

      • Regular verification of strain identity

    • For recombinant systems:

      • Sequence verification of all constructs

      • Analysis of plasmid stability

      • Consistent selection pressure maintenance

  • Growth condition standardization

    • Control critical parameters:

      • Media composition (defined media preferred)

      • Temperature monitoring with calibrated equipment

      • Consistent inoculation procedures (standardized starting OD)

      • Growth phase standardization (harvest at specific OD values)

      • Controlled aeration and agitation rates

Technical reproducibility:

  • Equipment calibration and validation

    • Implement regular calibration protocols:

      • Document all calibration procedures and schedules

      • Use reference standards appropriate for each instrument

      • Maintain calibration logs and certificates

    • Perform system suitability tests before critical experiments

    • Include internal controls for instrument performance

  • Reagent quality control

    • Establish reagent validation procedures:

      • Verify critical reagent activity before use

      • Prepare master stocks with aliquoting to minimize freeze-thaw

      • Document lot-to-lot variation testing

      • Implement expiration date tracking

Data analysis reproducibility:

  • Standardized data processing

    • Document all data analysis procedures:

      • Raw data preprocessing steps

      • Algorithm selection with justification

      • Parameter settings and thresholds

      • Software versions and computational environment

    • Implement automated analysis pipelines when possible

    • Archive raw data in non-proprietary formats

  • Transparent reporting

    • Follow field-specific reporting guidelines

    • Report all experimental conditions that might affect outcomes

    • Document all outliers and how they were handled

    • Disclose all statistical tests performed (including unsuccessful analyses)

Contradiction handling:

  • Structured contradiction analysis

    • Apply the (α, β, θ) notation to classify and resolve contradictions :

      • Systematically document contradictory results

      • Identify minimal set of variables explaining contradictions

      • Design targeted experiments to resolve discrepancies

    • Create contradiction resolution decision trees

    • Maintain a laboratory "contradictions log" to track resolution progress

Reproducibility assessment table:

Reproducibility DomainImplementation StrategyValidation MethodAcceptance Criteria
Biological MaterialSequence verification of all constructsSanger sequencing100% match to reference sequence
Growth ConditionsStandardized media and conditionsGrowth curve analysisCoefficient of variation <10%
Protein PurificationDetailed SOP with critical parametersPurity and yield assessmentPurity >90%, yield variation <15%
Functional AssaysCalibration standards for each assayControl sample performanceControls within 2 SD of historical means
Data AnalysisScripted analysis pipelineIndependent analysis verification<5% difference in final results
Contradiction ResolutionSystematic testing of variablesControlled experimentsClear identification of variable causing contradiction

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.