M.Pneumoniae P30

Mycoplasma Pneumoniae P30 Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Roles in Pathogenesis

P30 is indispensable for M. pneumoniae virulence through two mechanisms:

  • Adhesion: Binds sialoglycoproteins and sulfated glycolipids on host respiratory epithelial cells . Mutants lacking P30 fail to adhere and become non-infectious .

  • Gliding Motility: Coordinates with P1 adhesin to enable movement along cilia, facilitating colonization .

Key Findings:

  • P30 stabilizes the P65 protein, maintaining structural integrity of the attachment organelle .

  • Deletion of Domain III disrupts adhesion complex formation, even if P1 is present .

Genetic Diversity and Mutational Analysis

Sequencing of p30 genes from clinical isolates reveals variations linked to pathogen adaptability:

  • Four conserved substitutions (e.g., V80G, L108S) in 16/18 Indian asthma patients .

  • Two major subtypes: M-129 (subtype 1) and FH-Liu (subtype 2), with FH-Liu predominating in clinical samples .

Table 2: Clinically Observed P30 Mutations

MutationAmino Acid ChangePrevalence (%)Functional Consequence
239A→GValine→Glycine89Altered receptor affinity
583C→TProline→Serine89Reduced antibody recognition

Diagnostic Applications

P30’s immunogenic C-terminal region is leveraged in serological assays:

  • ELISA Sensitivity: 78.57% sensitivity and 89.47% specificity when paired with recombinant P30B (amino acids 17–274) .

  • Multiplex Assays: Combined with P1 and P116 adhesins, P30 improves diagnostic accuracy .

Vaccine Development Challenges and Advances

Live-Attenuated Vaccines:

  • P30 mutants induced severe Th17-mediated inflammation in mice, limiting utility .
    Recombinant Subunit Vaccines:

  • Fusion of P30 (aa 17–274) with P1’s C-terminal (aa 1287–1518) elicited protective IgA in guinea pigs .

Table 3: P30-Based Vaccine Candidates

TypeAntigen DesignOutcomeLimitation
Live-attenuatedP30 deletion mutantAvirulent but caused Th17 pathologyUnsafe for clinical use
Recombinant fusionP30 + P1 C-terminalInduced mucosal IgA in guinea pigsRequires adjuvant optimization

Recombinant P30 Production

  • Expression Systems: E. coli-produced P30 with a C-terminal His-tag (18–19 kDa) achieves >95% purity .

  • Applications: Used in immunoassays and structural studies to map epitopes .

Product Specs

Introduction

Mycoplasma pneumonia, a type of atypical pneumonia, is caused by the bacterium M. pneumoniae. This infection primarily affects individuals under 40 years old, accounting for 15% to 50% of pneumonia cases in adults and even more in school-aged children. While those in crowded environments like schools and shelters are at higher risk, many patients lack identifiable risk factors. The membrane proteins P1, P30, and P116 of M. pneumoniae are recognized as adhesion factors, with P1 being the primary protein involved in colonization.

Description

Recombinant M.Pneumoniae P30, produced in E.Coli, is a non-glycosylated polypeptide chain with a molecular weight of 18-19kDa. It features a 6 amino acid His-tag fused at the C-terminus and is purified using proprietary chromatographic techniques.

Physical Appearance

The product appears as a clear, sterile-filtered solution.

Formulation

The MP-P30c solution is formulated with 25mM K2CO3, 0.025% NaN3, and PBS.

Stability

While the Recombinant MP-P30c protein remains stable at 4°C for up to one week, long-term storage below -18°C is recommended. The addition of a carrier protein (0.1% HSA or BSA) is suggested for long-term storage. It is crucial to avoid repeated freeze-thaw cycles.

Purity

The protein's purity exceeds 95%, as determined by 10% PAGE (coomassie staining).

Applications

This product is suitable for immunoassay applications.

Source

Escherichia Coli.

Q&A

What is the M. pneumoniae P30 protein and what is its significance in bacterial pathogenesis?

M. pneumoniae P30 is a 30 kDa adhesin protein (275 amino acids) located at the tip of M. pneumoniae's terminal organelle. It is the second protein (after P1) identified to be associated with cell adhesion and virulence. P30 plays a crucial role in enabling the bacterium to adhere to respiratory epithelial cells, which is an essential step for infection. Additionally, P30 participates in the gliding movement of M. pneumoniae, allowing it to move from the tips of epithelial cilia in the bronchioles to the surface of host cells. Mutants lacking the P30 protein are defective in adhesion and motility and are avirulent, highlighting its importance in pathogenesis .

How does the P30 protein differ from other adhesin proteins in M. pneumoniae?

While P1 is the most extensively studied adhesin, P30 exhibits more structural similarity to P1 than other major adhesion proteins. The P30 protein is part of a complex adhesion apparatus that includes P1, P116, P65, auxiliary proteins P40 and P90, and high molecular weight proteins HMW1-3. P30 specifically functions in cell adhesion, proper cell development, stability of the P65 protein, and gliding motility. The unique feature of P30 is its transmembrane orientation with an intracytoplasmic N terminus and an exposed C terminus, along with distinctive repeat sequences at its carboxy end that are not present in other adhesins .

What are the main structural domains of the P30 protein?

The P30 protein contains several significant structural domains:

  • A transmembrane (TM) region that separates structural domains I and II

  • The TM structural domain includes a GxxxG sequence (G92 to G96) that facilitates interactions between TM helices

  • Three types of repeat sequences at its carboxy end:

    • One stretch of Pro-Gly-Met-Ala-Pro-Arg occurring seven times

    • Two stretches of Pro-Gly-Met-Pro-Pro-His repeating three times

    • Pro-Gly-Phe-Pro-Pro-Gln repeating three times
      Substitutions within structural domain II or deletions within structural domain III reduce motility and cell adhesion capabilities .

What techniques are most effective for determining the three-dimensional structure of P30 protein?

For high-resolution structural determination of P30 protein, researchers should consider:

  • X-ray crystallography: This requires successful crystallization of the purified protein, which can be challenging for membrane proteins like P30. The process would involve:

    • Expression of recombinant P30 protein (potentially using systems like the pMAL-p2x vector that has shown success)

    • Purification to homogeneity using affinity chromatography followed by gel filtration

    • Crystallization screening to identify optimal conditions

    • X-ray diffraction data collection and structure determination

  • Cryo-electron microscopy (cryo-EM): A powerful alternative for membrane proteins that:

    • Does not require protein crystallization

    • Can visualize the protein in a near-native state

    • May be combined with single-particle analysis for higher resolution

  • Nuclear Magnetic Resonance (NMR) spectroscopy: Particularly useful for analyzing specific domains or fragments of P30 .

The exact three-dimensional structure remains unresolved as of 2025, presenting an important research opportunity.

How can researchers effectively express and purify P30 protein for structural studies?

Based on documented experimental approaches:

  • Gene fragment selection:

    • Full-length P30 protein (825 bp) has shown poor expression levels

    • The P30B fragment (744 bp) demonstrates better expression

  • Expression system options:

    • pQE-30 vector system has shown moderate expression levels but produces insoluble protein

    • pMAL-p2x fusion vector system produces a soluble MBP-P30 fusion protein of approximately 87 kDa

  • Purification strategy:

    • For pQE-30 system: Purification under denaturing conditions using nickel-nitriloacetic acid affinity column

    • For pMAL-p2x system: Purification on an amylose column followed by SDS-PAGE and electroelution

Researchers should note that attempts to purify and refold the protein from the pQE-30 system under non-denaturing conditions have been unsuccessful. The pMAL-p2x system is preferable for obtaining soluble protein suitable for structural and functional studies .

What are the key sequence characteristics of the P30 gene that researchers should be aware of?

The P30 gene contains an open reading frame of 825 nucleotides coding for a protein of 275 amino acids with a calculated molecular mass of 29.743 kDa. Critical sequence features include:

  • A single UGA codon (nucleotides 46-48) that codes for tryptophan in M. pneumoniae rather than serving as a stop codon

  • Sequence variations observed in clinical isolates:

    • Some isolates match the reference strain M-129

    • Others show four base substitutions at positions 239, 323, 583, and 696

    • These substitutions result in three amino acid changes at positions 80 (valine-glycine), 108 (leucine-serine), and 195 (proline-serine)

    • One silent mutation at position 232 (glycine-glycine)

When designing primers for amplification, researchers should consider these variations and the unique codon usage. For expression in E. coli, it's advisable to design primers that exclude the UGA codon region to avoid premature termination of translation .

What is the mechanism by which P30 facilitates adhesion to host cells?

P30 participates in adhesion through multiple mechanisms:

  • Receptor recognition: P30 adheres to sialoglycoproteins and sulfated glycolipids on the surface of host cells

  • Coordinated function: P30 works in concert with the P1 adhesin, with both proteins sharing sequence homology

  • Structural positioning: Located at the tip of M. pneumoniae's terminal organelle, P30 is optimally positioned for initial contact with host cells

  • Signal transduction: Upon adhesion, P30 helps trigger changes in host cell metabolism and ultrastructure

The complete molecular mechanism is not yet fully elucidated, presenting an important area for further research. Studies suggest that the C-terminal region with its repeat sequences plays a significant role in the adhesion process, as it is exposed on the bacterial surface and can interact with host cell receptors .

How does P30 contribute to the gliding motility of M. pneumoniae?

P30 plays a crucial role in the gliding motility of M. pneumoniae through several mechanisms:

  • Terminal organelle architecture: P30 is essential for the proper development and functioning of the terminal organelle, which serves as the leading edge during gliding

  • Motility correlation: Experimental evidence shows that:

    • M. pneumoniae HA mutant II-3, which completely lacks the P30 protein, is non-motile

    • HA mutant II-7, which produces an altered P30 protein, shows 50-fold less motility than wild-type bacteria

  • Developmental impact: Mutations in P30 result in developmental defects that directly impact the gliding apparatus

These findings suggest that P30 may be involved in the mechanical aspects of motility, possibly by facilitating the attachment-detachment cycles necessary for gliding movement across surfaces. The transmembrane orientation of P30 may enable it to transduce forces between the cytoskeleton-like elements inside the cell and the external environment .

What experimental approaches can be used to study P30's role in M. pneumoniae virulence?

Several complementary approaches are recommended for studying P30's role in virulence:

  • Mutational analysis:

    • Generate specific mutations in different domains of P30

    • Assess the impact on adhesion, motility, and virulence

    • Compare with known mutants (e.g., class II non-cellular adhesion mutants)

  • Cell culture models:

    • Infect respiratory epithelial cell lines with wild-type and P30 mutant strains

    • Quantify adhesion efficiency, cytopathic effects, and inflammatory responses

    • Use fluorescently labeled bacteria to track adhesion and motility in real-time

  • Animal infection models:

    • Compare virulence of wild-type and P30 mutant strains in appropriate animal models

    • Assess bacterial loads, histopathology, and disease progression

  • Molecular interaction studies:

    • Identify host cell receptors that interact with P30

    • Characterize protein-protein or protein-glycan interactions using techniques like surface plasmon resonance

    • Perform competition assays with purified P30 protein or antibodies against P30 .

What PCR protocols are recommended for amplifying the P30 gene from clinical samples?

Based on successful experimental approaches, the following PCR protocol is recommended:

  • Primer design for full-length P30 gene (825 bp):

    • Forward primer targeting the 5' region of the gene

    • Reverse primer targeting the 3' region

    • Include appropriate restriction sites for subsequent cloning

  • PCR conditions:

    • Initial denaturation: 94°C for 5 minutes

    • 35 cycles of:

      • Denaturation: 94°C for 30 seconds

      • Annealing: 55-58°C for 30 seconds

      • Extension: 72°C for 1 minute

    • Final extension: 72°C for 10 minutes

  • DNA extraction considerations:

    • Direct extraction from clinical samples (throat swabs) is possible

    • Multiple independent amplifications (at least three) should be performed for each sample to confirm sequence variations

  • Sequence verification:

    • Sequence the amplified products in both directions

    • Compare with reference sequences (e.g., strain M-129, GenBank accession no. M57245)

    • Use appropriate software tools (e.g., Clustal W, Gene Doc) for sequence analysis

    • Use the Mycoplasma coding table for protein translation .

What are the optimal expression systems for producing recombinant P30 protein?

Based on experimental evidence, the following expression systems have been evaluated:

Expression SystemVectorHost CellProtein YieldSolubilityPurification MethodNotes
His-taggedpQE-30E. coli M-15ModerateInsolubleNi-NTA under denaturing conditionsFull-length P30 (P30A) shows poor expression
His-taggedpQE-30E. coli M-15ModerateInsolubleNi-NTA under denaturing conditionsP30B fragment shows better expression
MBP fusionpMAL-p2xE. coli TB-1GoodSolubleAmylose column followed by gel electrophoresis and electroelutionProduces ~87 kDa fusion protein with ~80% purity

The pMAL-p2x fusion system is currently the most promising approach, as it:

  • Produces soluble protein, avoiding refolding challenges

  • Results in higher expression levels

  • Yields protein that maintains immunological recognition by anti-M. pneumoniae antibodies

  • Enables recognition by M. pneumoniae-infected patient sera in immunoblots

When designing expression constructs, researchers should note that excluding the UGA codon region (which codes for tryptophan in M. pneumoniae but is a stop codon in E. coli) improves expression success .

How can researchers develop effective immunoassays for detecting P30 antibodies in clinical samples?

Based on documented approaches, the following steps are recommended for developing P30-based immunoassays:

  • Antigen preparation:

    • Express the C-terminal region of P30 (P30B fragment) as an MBP fusion protein

    • Purify using affinity chromatography followed by gel electrophoresis if needed

    • Ensure protein quality through SDS-PAGE and Western blot verification

  • ELISA protocol development:

    • Coat microplate wells with purified P30B protein at optimized concentration

    • Block with appropriate blocking buffer (e.g., 5% non-fat milk)

    • Incubate with diluted patient sera (1:100 dilution has shown good results)

    • Detect with enzyme-conjugated anti-human IgG

    • Develop with appropriate substrate and measure optical density

  • Assay validation:

    • Determine sensitivity and specificity against gold standard methods

    • Establish appropriate cutoff values based on ROC curve analysis

    • Include positive and negative controls in each assay run

  • Performance enhancement:

    • Consider using a combination of P30 with other adhesin proteins (e.g., P1)

    • Optimize buffer conditions, incubation times, and washing steps

    • Validate with a diverse set of clinical samples

Previous studies have demonstrated a sensitivity of 78.57% and specificity of 89.47% for P30B-based ELISA compared to commercial kits, suggesting that P30 can be effective as part of a multi-antigen approach to M. pneumoniae diagnosis .

What are the known genetic variations in the P30 gene across different M. pneumoniae isolates?

Research on P30 gene sequences from clinical isolates has revealed significant genetic diversity:

  • Reference strain variations:

    • Strain M-129 sequence (GenBank accession no. M57245) serves as a reference

    • Strain FH-Liu shows consistent variations from M-129

  • Clinical sample variations:

    • In a study of 18 Indian clinical isolates, 16 samples (89%) showed four base substitutions compared to strain M-129:

      • Position 239: Leading to valine-glycine amino acid change at position 80

      • Position 323: Leading to leucine-serine amino acid change at position 108

      • Position 583: Leading to proline-serine amino acid change at position 195

      • Position 696: Silent mutation (glycine-glycine) at position 232

  • Functional implications:

    • The amino acid substitutions occur in regions that may affect protein structure and function

    • The valine-glycine substitution at position 80 is near the transmembrane domain

    • The proline-serine substitution at position 195 may alter the conformation of the protein

These variations should be considered when designing diagnostic tests, vaccines, or therapeutic interventions targeting P30 .

How do mutations in the P30 gene affect M. pneumoniae adherence and motility?

Studies of M. pneumoniae mutants have provided important insights into the relationship between P30 mutations and bacterial function:

  • Complete P30 deficiency:

    • M. pneumoniae HA mutant II-3 completely lacks P30 protein

    • This mutant is non-motile and defective in cell adhesion

    • It also shows developmental defects in the terminal organelle

  • Altered P30 protein:

    • M. pneumoniae HA mutant II-7 produces an altered P30 protein

    • This mutant exhibits approximately 50-fold less motility than wild-type bacteria

    • It retains some adhesion capability but at significantly reduced levels

  • Domain-specific effects:

    • Substitutions within structural domain II of P30 reduce motility and cell adhesion

    • Deletions within structural domain III similarly affect these functions

    • Mutations in the GxxxG sequence (G92 to G96) in the transmembrane domain affect the distribution of P30 across the M. pneumoniae membrane

These findings demonstrate that different regions of the P30 protein contribute specifically to adherence and motility functions, and that even partial alterations can significantly impact bacterial virulence .

What advanced methodologies can be used to study the effects of P30 mutations on protein function?

Several sophisticated approaches can be employed to investigate the functional impact of P30 mutations:

  • Site-directed mutagenesis:

    • Create precise mutations in specific domains of P30

    • Express these mutants in M. pneumoniae using genetic transformation techniques

    • Assess the effects on adhesion, motility, and cell development

  • Complementation studies:

    • Introduce wild-type or mutant P30 genes into P30-deficient strains

    • Evaluate the restoration of function to determine critical regions

    • Compare natural variants to engineered mutations

  • Protein-protein interaction analysis:

    • Use techniques like bacterial two-hybrid systems or co-immunoprecipitation

    • Identify interactions between P30 and other terminal organelle proteins

    • Determine how mutations affect these interactions

  • Live-cell imaging:

    • Employ fluorescent protein fusions to visualize P30 localization

    • Track the dynamics of wild-type versus mutant P30 during gliding motility

    • Use high-resolution microscopy techniques like TIRF or super-resolution microscopy

  • Structure-function correlation:

    • Combine structural data (when available) with functional assays

    • Map mutations onto the three-dimensional structure

    • Predict functional effects based on structural changes .

How does the P30 protein induce immune responses during M. pneumoniae infection?

The P30 protein is highly immunogenic and induces significant immune responses during infection:

  • Antibody responses:

    • P30 elicits production of specific antibodies during natural infection

    • These antibodies can be detected in the serum of infected individuals

    • The C-terminal region with its repeat sequences appears particularly immunogenic

  • Epitope recognition:

    • The exposed C-terminus of P30 contains important B-cell epitopes

    • The proline-rich repeat regions at the carboxy end are likely immunodominant

    • Both linear and conformational epitopes may be present

  • Kinetics of immune response:

    • Anti-P30 antibodies appear relatively early in infection

    • Both IgM and IgG antibodies against P30 can be detected

    • The antibody response to P30 may persist for months after infection

  • Cross-reactivity considerations:

    • Some epitopes may show homology with P30-like proteins from other Mycoplasma species

    • Potential cross-reactivity with M. gallisepticum MGC2 protein and M. genitalium P32 protein

The strong immunogenicity of P30 makes it a valuable candidate for diagnostic applications and vaccine development, though the regulatory mechanisms of immune responses induced by P30 require further investigation .

What are the most sensitive methods for detecting P30 expression in clinical or laboratory samples?

Multiple methods can be employed for detecting P30 expression, each with specific advantages:

  • Molecular detection methods:

    • RT-PCR for detecting P30 mRNA expression

    • Real-time quantitative PCR for quantifying expression levels

    • Next-generation sequencing for comprehensive gene expression analysis

  • Protein detection methods:

    • Western blotting using specific anti-P30 antibodies

    • Immunofluorescence microscopy for localization studies

    • Flow cytometry for quantitative analysis of surface expression

  • Mass spectrometry approaches:

    • Liquid chromatography-tandem mass spectrometry (LC-MS/MS)

    • Multiple reaction monitoring (MRM) for targeted quantification

    • MALDI-TOF for rapid identification

  • Enhanced sensitivity techniques:

    • Signal amplification methods (e.g., tyramide signal amplification)

    • Digital PCR for absolute quantification

    • Proximity ligation assay for protein detection in situ

The choice of method depends on the research question, available sample types, and required sensitivity. For clinical samples with potentially low bacterial loads, combining molecular detection of the P30 gene with immunological detection of the protein provides the most comprehensive assessment .

How can P30 be utilized in developing vaccines against M. pneumoniae?

P30 offers significant potential as a vaccine candidate through several approaches:

  • Subunit vaccine development:

    • Express recombinant P30 protein or fragments (particularly the immunogenic C-terminal region)

    • Formulate with appropriate adjuvants to enhance immunogenicity

    • Evaluate antibody responses and protective efficacy in animal models

  • Multi-antigenic vaccine approaches:

    • Combine P30 with other adhesin proteins (e.g., P1)

    • This combination might provide broader protection against different strains

    • Could address potential genetic variations in either protein

  • DNA vaccine strategy:

    • Construct plasmids encoding the P30 gene

    • Optimize codon usage for enhanced expression in mammalian cells

    • Deliver via appropriate routes to induce both humoral and cellular immunity

  • Epitope-based vaccine design:

    • Identify specific protective epitopes within P30

    • Create synthetic peptides or epitope-presenting constructs

    • Target both B-cell and T-cell epitopes for comprehensive immunity

  • Delivery system considerations:

    • Evaluate different adjuvants for optimal immune stimulation

    • Consider mucosally administered vaccines for respiratory pathogens

    • Explore nanoparticle or virus-like particle platforms for epitope presentation

When developing P30-based vaccines, researchers must account for sequence variations across clinical isolates and ensure that vaccine constructs induce antibodies capable of blocking adhesion and neutralizing the pathogen .

What are the current gaps in our understanding of P30 protein structure and function?

Despite significant progress, several important knowledge gaps remain:

What integration of omics approaches would advance P30 research?

A comprehensive multi-omics strategy would significantly enhance our understanding of P30:

  • Genomics integration:

    • Whole-genome sequencing of clinical isolates to correlate P30 variations with disease phenotypes

    • Comparative genomics to understand P30 evolution across Mycoplasma species

    • Metagenomics to study P30 in the context of the respiratory microbiome

  • Transcriptomics applications:

    • RNA-seq to analyze gene expression changes in host cells following exposure to wild-type vs. P30-mutant M. pneumoniae

    • Small RNA profiling to identify potential regulatory mechanisms of P30 expression

    • Single-cell transcriptomics to capture heterogeneity in bacterial and host cell responses

  • Proteomics approaches:

    • Interactomics to identify P30 protein-protein interactions within bacterial cells and with host proteins

    • Post-translational modification analysis of P30 and its impact on function

    • Quantitative proteomics to measure changes in the bacterial and host proteome during infection

  • Structural biology integration:

    • Cryo-EM of the terminal organelle to visualize P30 in its native context

    • Integrative modeling combining data from multiple structural techniques

    • Molecular dynamics simulations to understand P30 function in membranes

  • Systems biology framework:

    • Network analysis to position P30 within the broader context of bacterial pathogenesis

    • Predictive modeling of P30's role in adhesion and motility

    • Integration of multi-omics data to develop comprehensive models of M. pneumoniae pathogenesis .

How might innovative biotechnology techniques enhance P30 research and applications?

Cutting-edge biotechnology approaches offer promising avenues for advancing P30 research:

  • CRISPR-Cas systems for Mycoplasma:

    • Development of CRISPR-based genome editing for M. pneumoniae

    • Creation of precise P30 mutations or domain swaps

    • Generating reporter strains for high-throughput studies

  • Synthetic biology approaches:

    • Minimal genome studies to determine the essential nature of P30

    • Redesigning P30 with optimized or novel functions

    • Creating synthetic cellular systems to study P30 in isolation

  • Advanced imaging technologies:

    • Super-resolution microscopy to visualize P30 distribution and dynamics

    • Correlative light and electron microscopy for structural-functional studies

    • Live-cell imaging to track P30 during the infection process

  • Microfluidic systems:

    • Organ-on-chip models of respiratory epithelium for infection studies

    • Single-cell analysis of host-pathogen interactions

    • High-throughput screening of anti-adhesion compounds targeting P30

  • Therapeutic applications:

    • Development of P30-targeting antibodies or nanobodies

    • Peptide inhibitors designed to block P30-mediated adhesion

    • mRNA vaccines encoding optimized P30 antigens

  • Diagnostic innovations:

    • CRISPR-based detection systems for P30 gene variants

    • Nanobody-based rapid tests for P30 protein

    • Digital biomarker development for monitoring M. pneumoniae infections .

How can P30 research inform the development of novel antimicrobial strategies?

P30 research provides several promising avenues for antimicrobial development:

  • Adhesion inhibitors:

    • Design of peptides or small molecules that mimic host receptors to block P30-mediated attachment

    • Development of antibodies or nanobodies targeting the adhesion domains of P30

    • Creation of glycomimetics to competitively inhibit P30 binding to sialoglycoproteins

  • Motility disruptors:

    • Compounds that interfere with the gliding motility function of P30

    • Targeting the interaction between P30 and other components of the motility apparatus

    • Disrupting the energy transfer mechanisms required for P30-mediated movement

  • Structure-based drug design:

    • Using the structural data (when available) to design specific inhibitors

    • Virtual screening of compound libraries against P30 binding sites

    • Fragment-based approaches to develop high-affinity ligands

  • Immunotherapeutic approaches:

    • Passive immunization with anti-P30 antibodies

    • Development of immunomodulators that enhance the host response to P30

    • Combination therapy with conventional antibiotics and anti-P30 agents

  • CRISPR-based antimicrobials:

    • Targeting the P30 gene with CRISPR-Cas systems delivered via appropriate vectors

    • Development of programmable nucleases specific for P30 gene sequences

    • Creating pressure for evolutionary trade-offs that reduce virulence .

What research models are most appropriate for studying P30 function in the context of respiratory infection?

Several complementary models can be employed to study P30 function:

  • Cell culture models:

    • Human respiratory epithelial cell lines (e.g., A549, BEAS-2B)

    • Primary human bronchial epithelial cells

    • Air-liquid interface cultures that recapitulate the respiratory epithelium

    • Advantages: Human-relevant, controlled conditions, amenable to high-throughput studies

    • Limitations: Lack systemic immune components, simplified compared to in vivo

  • Animal models:

    • Mouse models of M. pneumoniae infection

    • Syrian hamster model (more susceptible to M. pneumoniae)

    • Non-human primate models for advanced studies

    • Advantages: Complete immune system, physiological relevance

    • Limitations: Species differences in receptor distribution, ethical considerations

  • Organoid models:

    • Lung organoids derived from stem cells

    • Respiratory tract organoids containing multiple cell types

    • Advantages: Human-relevant, 3D architecture, multiple cell types

    • Limitations: Lack systemic components, technical challenges

  • Microfluidic "organ-on-chip" systems:

    • Lung-on-chip models incorporating flow and mechanical forces

    • Multi-organ systems to study extrapulmonary complications

    • Advantages: Control over microenvironment, incorporation of physical forces

    • Limitations: Technical complexity, validation requirements

  • Mathematical and computational models:

    • Agent-based models of M. pneumoniae adhesion and motility

    • Systems biology approaches to model host-pathogen interactions

    • Advantages: Hypothesis generation, integration of diverse data types

    • Limitations: Require experimental validation .

How can researchers design experiments to assess the impact of P30 genetic variation on vaccine efficacy?

A comprehensive experimental design to evaluate the impact of P30 genetic variation on vaccine efficacy would include:

  • Sequence analysis phase:

    • Collect and sequence P30 genes from a diverse set of clinical isolates

    • Identify major variant groups and representative strains

    • Perform epitope prediction and conservation analysis across variants

  • Vaccine construct development:

    • Design multiple vaccine candidates:
      a. Full-length P30 from reference strain
      b. Conserved region-focused constructs
      c. Multi-variant constructs incorporating epitopes from different variants
      d. Consensus sequence-based constructs

  • Immunogenicity testing:

    • Evaluate antibody responses in animal models

    • Assess cross-reactivity against different P30 variants using ELISA

    • Perform epitope mapping to identify immunodominant regions

    • Measure neutralizing antibody titers against different variants

  • Functional assays:

    • Adhesion inhibition assays using sera from vaccinated animals

    • Flow cytometry to assess antibody binding to different M. pneumoniae strains

    • Complement-dependent bactericidal activity against variant strains

  • Challenge studies:

    • Test protection against challenge with different variant strains

    • Measure bacterial loads, histopathology, and clinical parameters

    • Analyze correlates of protection across variant challenges

  • Adaptive vaccine design:

    • Utilize results to refine vaccine constructs

    • Consider prime-boost strategies with different variants

    • Develop polyvalent formulations if necessary

This experimental approach would provide comprehensive data on the impact of P30 variation on vaccine efficacy and guide the development of broadly protective vaccines .

Product Science Overview

Introduction

Mycoplasma pneumoniae is a unique, cell wall-deficient bacterium that primarily infects the human respiratory tract. It is a significant cause of respiratory infections, including tracheobronchitis and atypical pneumonia, often referred to as “walking pneumonia.” The bacterium is particularly prevalent among school-aged children and young adults .

The Role of P30 Protein

The P30 protein is one of the key surface proteins of Mycoplasma pneumoniae. It is associated with the bacterium’s terminal organelle, which plays a crucial role in its ability to adhere to host cells (cytadherence) and in gliding motility . The terminal organelle is a specialized structure that allows the bacterium to attach to the respiratory epithelium, facilitating colonization and infection .

Importance of P30 in Cytadherence and Motility

Research has shown that the P30 protein is essential for the bacterium’s adherence to host cells. Mutants lacking the P30 protein exhibit significant defects in cytadherence, which impairs their ability to colonize the host . Additionally, the P30 protein is involved in the gliding motility of Mycoplasma pneumoniae, a unique form of movement that allows the bacterium to navigate the mucosal surfaces of the respiratory tract .

Recombinant P30 Protein

The recombinant P30 protein is a genetically engineered version of the native P30 protein. It is produced using recombinant DNA technology, which involves inserting the gene encoding the P30 protein into a suitable expression system, such as bacteria or yeast, to produce the protein in large quantities . This recombinant protein can be used for various research and diagnostic purposes.

Applications in Research and Diagnostics

The recombinant P30 protein is valuable in studying the pathogenesis of Mycoplasma pneumoniae infections. It allows researchers to investigate the specific functions of the P30 protein in cytadherence and motility, as well as its interactions with other proteins and host cell receptors . Additionally, the recombinant P30 protein can be used in serological assays to detect antibodies against Mycoplasma pneumoniae in patient samples, aiding in the diagnosis of infections .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.