M.Pneumoniae P1-C

Mycoplasma Pneumoniae P1-C Recombinant
Shipped with Ice Packs
In Stock

Description

Functional Role in Pathogenesis

P1-C is essential for M. pneumoniae pathogenicity:

  • Adhesion: Antibodies targeting P1-C reduce bacterial adherence to host cells by 56–75% in vitro .

  • Immune evasion: Genetic variability in other P1 regions (e.g., N-terminal) diverts immune responses, while P1-C’s conservation allows sustained infectivity .

  • Cytoskeletal interaction: Collaborates with proteins like HMW1–HMW3 and P30 to stabilize the attachment organelle .

Antibody Inhibition Studies

Polyclonal antibodies against P1-C significantly impair M. pneumoniae adhesion:

Antibody TargetReduction in AdherenceSignificance (p-value)
P1-C (Lys1376–Asp1521)56%9×1069 \times 10^{-6}
P1-C (Ala1400–Asp1521)75%1×1061 \times 10^{-6}

Genetic Variability and Clinical Impact

  • P1 genotypes: M. pneumoniae strains are classified into P1-1 and P1-2 based on RepMP2/3/4 repeats in the P1 gene .

  • Disease severity: P1-2 strains correlate with higher CRP levels (p<0.05p < 0.05) and hospitalization rates (OR: 1.83) .

Diagnostic and Therapeutic Applications

  • Serological assays: Recombinant P1-C (expressed in E. coli) is used in ELISA to detect IgG antibodies in 74–90% of infected patients .

  • Vaccine development: P1-C’s conserved epitopes make it a candidate for subunit vaccines .

  • Monoclonal antibodies: Clone 6533 targets P1-C for diagnostic capture assays .

Clinical and Epidemiological Insights

  • Geographic distribution: P1-2 strains dominate in recent outbreaks, linked to severe pediatric lower respiratory tract infections .

  • Immune response: Anti-P1-C IgG titers peak during convalescence, suggesting role in long-term immunity .

Future Directions

  • Structural optimization: Cryo-EM and X-ray crystallography data (e.g., PDB 6T6U) are refining P1-C’s conformational epitopes .

  • Multivalent vaccines: Combining P1-C with P40/P90 antigens may enhance efficacy .

Product Specs

Introduction
Mycoplasma pneumonia, a type of atypical pneumonia, is caused by the bacterium M. pneumoniae and primarily affects individuals under 40. This disease represents 15-50% of pneumonia cases in adults, particularly school-aged children. Individuals in crowded environments like schools and shelters are at higher risk, though many cases have no clear risk factor. Key to the bacteria's colonization are adhesin proteins, notably P1, P30, and P116, found on the membrane. P1 plays a crucial role in this process.
Description
This recombinant Mycoplasma Pneumoniae protein represents the C-terminal region (P1C) of the P1 protein. Expressed in E. coli, it encompasses 362 amino acids and includes a 6-His tag. This peptide serves as an antigen for diagnosing M. pneumoniae infections.
Physical Appearance
Clear, sterile-filtered solution.
Formulation
The Mycoplasma Pneumoniae P1-C is supplied in 1x PBS with a pH of 7.4.
Stability
Store the product at -20°C upon receipt. Avoid repeated freeze-thaw cycles.
Purity
Purity exceeds 95% as assessed by 12% SDS-PAGE and Coomassie blue staining.
Applications

Suitable for immunoassays.

Source
E.Coli
Purification Method
The recombinant fusion protein was purified by GSH affinity chromatography technique.

Q&A

What is the M. pneumoniae P1 adhesin and why is its C-terminal region significant?

The P1 adhesin is a critical virulence factor of Mycoplasma pneumoniae that mediates attachment to host cells. The C-terminal region (P1-C) is particularly significant because it contains immunodominant epitopes that provoke robust immune responses. Research has demonstrated that the C-terminal part of the P1 protein (amino acid residues 1160 to 1521) is highly immunogenic, showing strong reactivity with M. pneumoniae IgG antibody-positive patient sera on immunoblotting . The P1-C region is not only important for immunological recognition but also plays a crucial role in cytadherence to respiratory epithelial cells. This region has been successfully expressed in E. coli and shown to have potential for diagnostic applications and vaccine development .

How does the genomic structure of the P1 gene affect expression in laboratory systems?

The expression of M. pneumoniae P1 protein in heterologous systems faces a unique challenge due to the unusual codon usage in mycoplasmas. In M. pneumoniae, the UGA codon encodes tryptophan instead of functioning as a stop codon as it does in standard bacterial expression systems like E. coli . This difference leads to premature termination when attempting to express full-length P1 protein in conventional expression systems.

Researchers have overcome this limitation through several strategies:

  • Expressing specific regions of the P1 gene that lack UGA codons

  • Site-directed mutagenesis to change UGA to UGG codons

  • PCR amplification with primers designed to modify UGA codons to UGG

For example, in the expression of the P1-C1 region, researchers used primers designed to change the UGA codon to UGG for the E. coli expression system: forward (P-1e) 5′-AGA TCT GAA TTC GCG GCC TTT CGT GGC AGT TGG GTC-3′ and reverse (P-If) 5′-CAT TGG CTG CAG ATC AGG CCA CTG GTT AAA CGG ACT AAA CAA-3′ .

How do P1-1 and P1-2 genotypes differ in their clinical implications?

Research has revealed significant clinical differences between infections caused by M. pneumoniae strains with different P1 genotypes:

ParameterP1-1 GenotypeP1-2 GenotypeStatistical Significance
Baseline C-reactive proteinLower median levelsHigher median levelsSignificant
Hospital admission rateLowerHigherSignificant
Disease severityModerateMore severeSignificant

Children infected with P1-2 strains presented with significantly higher median baseline C-reactive protein levels and required hospital admission more frequently than those with P1-1 infections . The P1 genotype had significant predictive value in multiple linear regression models forecasting C-reactive protein levels and significantly affected the likelihood of hospital admission in logistic regression analyses . These findings suggest that the two M. pneumoniae P1 genotypes may have different pathogenic potential, with P1-2 strains possibly causing a more severe course of lower respiratory tract infections.

What are the optimal methods for expressing recombinant P1-C protein?

Based on the research literature, the following methodological approach has proven effective for expressing the C-terminal region of P1 protein:

  • PCR Amplification:

    • Design primers that change UGA codons to UGG

    • Use genomic DNA from M. pneumoniae as template

    • For P1-C1 region: Use forward primer 5′-AGA TCT GAA TTC GCG GCC TTT CGT GGC AGT TGG GTC-3′ and reverse primer 5′-CAT TGG CTG CAG ATC AGG CCA CTG GTT AAA CGG ACT AAA CAA-3′

  • Cloning Strategy:

    • Initial cloning into pGEMT vector for sequence verification

    • Restriction digestion with appropriate enzymes (BglII and PstI for P1-C1)

    • Subcloning into expression vector pQE-30

  • Expression Conditions:

    • Transform M15 E. coli cells with recombinant plasmid

    • Cultivate in LB broth with appropriate antibiotics (ampicillin 100 μg/ml, kanamycin 25 μg/ml)

    • Grow at 37°C until OD reaches 0.5-0.6

    • Induce protein expression with 1 mM IPTG

    • Continue incubation for 3 hours at 37°C with shaking

This methodology has successfully yielded expressed P1-C1 protein that demonstrates immunoreactivity with patient sera, confirming its potential utility in diagnostic applications.

How can researchers effectively purify and characterize recombinant P1-C protein?

Purification and characterization of recombinant P1-C protein involve several key steps:

  • Protein Purification:

    • Harvest bacterial cells by centrifugation

    • Lyse cells under denaturing conditions (8M urea)

    • Purify using Ni-NTA affinity chromatography (leveraging the His-tag from the pQE-30 vector)

    • Elute with imidazole-containing buffer

    • Dialyze against phosphate-buffered saline to remove urea

  • Characterization Methods:

    • SDS-PAGE: Confirm protein size and purity

    • Western Blotting:

      • Probe with Penta-His monoclonal antibodies (1:2,000 dilution) to confirm expression

      • Test with anti-M. pneumoniae antibody-positive patient sera (1:100 dilution) to confirm immunogenicity

    • ELISA: Compare reactivity with commercial kits to validate diagnostic potential

When implementing these methods, researchers observed that the P1-C1 protein exhibited strong immunoreactivity with patient sera, whereas the N-terminal region (P1-N1) showed no reactivity despite previous reports of its immunogenicity in peptide form. This suggests that the conformation of expressed proteins may affect epitope accessibility and immunoreactivity.

What immunological assays are most effective for evaluating P1-C immune responses?

Based on current research, several immunological assays have proven effective for evaluating immune responses to P1-C:

  • Antibody Detection:

    • Western blotting with patient sera to evaluate immunoreactivity

    • ELISA using purified P1-C as coating antigen to quantify antibody responses

    • Comparison with commercial serological assays for validation

  • Cytokine Profiling:

    • Measurement of inflammatory cytokines (IL-6) and immunomodulatory cytokines (IL-4, IL-10, IFN-γ) to assess immune response patterns

    • Analysis of cytokine levels in lung tissue or bronchoalveolar lavage fluid in animal models

  • Cellular Immunity Assessment:

    • T-cell proliferation assays in response to P1-C stimulation

    • Evaluation of CD4+ and CD8+ T-cell responses by flow cytometry

    • Measurement of T-cell cytokine production profiles

In vaccine studies, researchers have observed that P1-C-based mRNA vaccines elicited potent humoral and cellular immune responses, effectively reducing inflammation. These vaccines notably decreased IL-6 levels in the lungs of infected mice while concurrently elevating IL-4, IL-10, and IFN-γ levels post-immunization .

How can P1-C be utilized in developing mRNA vaccines against M. pneumoniae?

Development of mRNA vaccines targeting M. pneumoniae P1-C represents a promising avenue for preventing mycoplasma infections. The following methodological approach has shown success:

  • Target Selection:

    • Focus on the C-terminal region of P1 adhesin, which contains immunodominant epitopes

    • Specifically target amino acid residues 1160 to 1521, which have demonstrated strong immunogenicity

  • mRNA Vaccine Design:

    • Design mRNA encoding the P1-C region with optimized codons for mammalian expression

    • Incorporate modifications to enhance stability and translation efficiency

    • Encapsulate in lipid nanoparticles to protect mRNA and facilitate cellular uptake

  • Immunization Protocol:

    • Administer via intramuscular injection in a prime-boost regimen

    • Monitor both humoral (antibody) and cellular immune responses

    • Challenge with live M. pneumoniae to assess protective efficacy

Recent research has demonstrated that an mRNA vaccine targeting the P1 adhesin elicited potent humoral and cellular immune responses in BALB/c mice. The vaccine effectively diminished inflammation, reduced IL-6 levels in the lungs of infected mice, and elevated IL-4, IL-10, and IFN-γ levels post-immunization. Furthermore, the vaccine reduced pathological changes in the lungs and decreased M. pneumoniae DNA copy numbers in infected animals .

What are the potential advantages of P1-C-based diagnostics over conventional methods?

P1-C-based diagnostic methods offer several advantages over conventional diagnostic approaches for M. pneumoniae:

Diagnostic MethodAdvantagesLimitations
P1-C-based ELISA- Higher specificity due to use of recombinant antigen
- Improved sensitivity over crude lysate assays
- Ability to distinguish between acute and past infections
- Requires specialized reagents
- May need validation against gold standards
Culture- Definitive identification
- Allows antibiotic susceptibility testing
- Time-consuming (2-4 weeks)
- Low sensitivity
- Technically demanding
PCR- Rapid results
- High sensitivity
- Detection of macrolide resistance
- Cannot distinguish viable from non-viable organisms
- Possible contamination issues
Complement Fixation- Established methodology
- Predominantly measures IgM
- Lower sensitivity
- Cannot differentiate between acute and past infection reliably

The development of recombinant P1-C antigens has enabled better-performing immunoassays for both antigen and antibody detection specific to M. pneumoniae . Research has demonstrated that a P1-enriched antigen increases the sensitivity and specificity of serologic diagnosis compared to traditional methods using partially purified lysates .

How do mutations in the P1-C region affect bacterial virulence and vaccine efficacy?

Mutations in the P1-C region can significantly impact bacterial virulence and vaccine efficacy through several mechanisms:

  • Virulence Implications:

    • Alterations in key adhesin epitopes may affect binding to host cell receptors

    • Different P1 genotypes (P1-1 and P1-2) demonstrate varying disease severity, with P1-2 associated with higher C-reactive protein levels and increased hospitalization rates

    • Mutations may affect immune evasion capabilities and persistence in the host

  • Vaccine Efficacy Considerations:

    • Vaccines targeting specific P1-C epitopes may have reduced efficacy against variant strains

    • Strain-specific mutations could lead to immune escape

    • Broader coverage may require inclusion of epitopes from multiple P1 variants

  • Research Approaches:

    • Comparative genomic analysis of P1 sequences from clinical isolates

    • Analysis of epitope conservation across strains

    • Assessment of cross-protection in animal models using heterologous challenge strains

    • Development of multivalent vaccines incorporating P1-C regions from different genotypes

Understanding the impact of P1-C mutations on virulence and vaccine efficacy requires ongoing surveillance of circulating strains and adaptation of vaccine formulations to address emerging variants, similar to approaches used for other variable pathogens.

What is the correlation between P1-C antibody titers and clinical outcomes?

Understanding the relationship between P1-C antibody titers and clinical outcomes provides valuable insights for both diagnostic and prognostic applications:

  • Antibody Response Patterns:

    • Patients with M. pneumoniae infections develop antibodies against P1-C that can be detected by immunoblotting and ELISA

    • The C-terminal region of P1 appears to be more immunogenic than the N-terminal region in natural infections

    • Antibody responses to P1-C may correlate with protection against reinfection

  • Clinical Correlations:

    • Higher antibody titers against P1-C may be associated with more effective clearance of infection

    • The quality and specificity of anti-P1-C antibodies may be more important than absolute titer

    • Differences in antibody responses between P1-1 and P1-2 infections might contribute to the observed differences in disease severity

  • Research Approaches:

    • Longitudinal studies measuring anti-P1-C antibody titers during acute infection and convalescence

    • Correlation of antibody levels with clinical parameters (hospitalization duration, oxygen requirement, radiological findings)

    • Assessment of neutralizing activity of anti-P1-C antibodies in vitro

Further research is needed to establish definitive correlations between specific antibody profiles and clinical outcomes, which could inform both diagnostic test interpretation and vaccine development strategies.

How can P1-C research inform treatment approaches for macrolide-resistant M. pneumoniae?

The emergence of macrolide-resistant and multidrug-resistant strains of M. pneumoniae presents significant challenges to clinical management . P1-C research offers several potential avenues for addressing this growing problem:

The successful development of an mRNA vaccine targeting P1 adhesin demonstrates the potential for vaccination as a strategy to prevent M. pneumoniae infections regardless of antibiotic susceptibility patterns, offering a promising approach to combat the increasing prevalence of antibiotic-resistant strains .

What are the current gaps in P1-C research and potential future directions?

Despite significant advances in understanding M. pneumoniae P1-C, several important knowledge gaps remain:

  • Structural Understanding:

    • Detailed three-dimensional structure of the P1-C region remains incompletely characterized

    • Structural basis for differential virulence between P1-1 and P1-2 genotypes

    • Conformational epitopes that may be critical for protective immunity

  • Host-Pathogen Interactions:

    • Molecular mechanisms by which P1-C interacts with specific host receptors

    • Host factors that determine susceptibility to infection and disease severity

    • Immune evasion mechanisms related to P1-C variability

  • Vaccine Optimization:

    • Optimal epitope selection for broad protection against diverse strains

    • Duration of immunity following P1-C-based vaccination

    • Correlates of protection against M. pneumoniae infection

  • Proposed Research Approaches:

    • Application of cryo-electron microscopy to resolve P1-C structure

    • Systems biology approaches to understand host response networks

    • Large-scale clinical trials of P1-C-based vaccines with long-term follow-up

    • Comparative genomics of P1 sequences from global surveillance

Addressing these knowledge gaps will require multidisciplinary approaches combining structural biology, immunology, microbiology, and clinical research to fully leverage P1-C as a target for diagnosis, treatment, and prevention of M. pneumoniae infections.

Product Science Overview

P1 Adhesin Protein

The P1 adhesin protein is one of the major surface proteins of Mycoplasma pneumoniae. It plays a crucial role in the bacterium’s ability to adhere to the respiratory epithelium of the human host. The P1 protein is part of a complex tip-like attachment organelle that includes several other proteins such as P30, P116, HMW1, HMW2, and HMW3 .

Recombinant P1-C Protein

The recombinant P1-C protein refers to a recombinant version of the C-terminal region of the P1 adhesin protein. This recombinant protein is expressed in Escherichia coli (E. coli) and contains 362 amino acids. It is often fused to a 6 His Tag to facilitate purification and detection .

Importance in Diagnosis

The recombinant P1-C protein has been identified as an immunodominant antigen in Mycoplasma pneumoniae. It is used in various serological assays, such as enzyme-linked immunosorbent assays (ELISA), to detect antibodies against Mycoplasma pneumoniae in patient serum samples. The use of specific antigens like the recombinant P1-C protein helps in distinguishing between current and previous infections, providing a more accurate diagnosis .

Research and Applications

Research has shown that the combination of recombinant P1-C protein with other antigens, such as the ATP synthase beta subunit (AtpD), can improve the performance of serological assays. This combination has been found to discriminate maximally between patients infected with Mycoplasma pneumoniae and healthy subjects, especially for the IgM class of antibodies .

The recombinant P1-C protein is also used in the development of point-of-care diagnostic assays, which are essential for the early and specific diagnosis of Mycoplasma pneumoniae infections. These assays are crucial for selecting the appropriate treatment, as Mycoplasma pneumoniae is innately resistant to β-lactam antibiotics, which are commonly used as the first-line treatment for RTIs .

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.