Recombinant Nocardia farcinica Pup--protein ligase (pafA) is a recombinant enzyme derived from the bacterium Nocardia farcinica. This enzyme is involved in the pupylation process, which is a post-translational modification similar to ubiquitination in eukaryotes. Pupylation is crucial for protein degradation and regulation in prokaryotes, particularly in mycobacteria and related species like Nocardia farcinica. The pafA enzyme specifically catalyzes the conjugation of the prokaryotic ubiquitin-like protein Pup to target proteins, marking them for degradation by the proteasome.
The mechanism of pafA involves a two-step reaction. First, it activates the C-terminal glutamate of Pup by forming a γ-glutamyl phosphate-mixed anhydride intermediate. This intermediate is then transferred to a lysine residue on the target protein, forming an isopeptide bond between Pup and the protein substrate . This process is essential for the regulation of protein turnover and is critical for the survival and virulence of pathogenic bacteria.
Recombinant Nocardia farcinica Pup--protein ligase (pafA) is commercially available, often produced in yeast or other expression systems. This availability facilitates research into the enzyme's structure, function, and potential applications.
| Feature | Recombinant Nocardia farcinica Pup--protein ligase (pafA) | Recombinant Mycobacterium tuberculosis Pup--protein ligase (pafA) |
|---|---|---|
| Source | Nocardia farcinica | Mycobacterium tuberculosis |
| Function | Conjugates Pup to target proteins for degradation | Essential for M. tuberculosis survival and virulence |
| Mechanism | Two-step reaction forming a γ-glutamyl phosphate intermediate | Similar two-step mechanism |
| Production | Often produced in yeast | Produced in E. coli |
| Applications | Research into protein degradation pathways | Potential drug target for tuberculosis |
Further research on Recombinant Nocardia farcinica Pup--protein ligase (pafA) could explore its structural and functional differences compared to other pafA enzymes, such as those from M. tuberculosis. This could lead to a deeper understanding of the pupylation pathway and its role in bacterial physiology, potentially uncovering new targets for antimicrobial therapies.
Recombinant Nocardia farcinica Pup Protein Ligase (pafA): Catalyzes the covalent attachment of the prokaryotic ubiquitin-like protein modifier Pup to proteasomal substrate proteins, thereby targeting them for proteasomal degradation. This process is known as pupylation. The ligation reaction involves the C-terminal glutamate of Pup and a substrate lysine residue.
KEGG: nfa:NFA_31700
STRING: 247156.nfa31700
Nocardia farcinica is a gram-positive, partially acid-fast, filamentous bacterium considered an opportunistic pathogen found in soil, decaying vegetation, and ventilation systems . It causes acute, subacute, or chronic infections known as nocardiosis, particularly in immunocompromised individuals. Patients at greatest risk include those with T-lymphocyte deficiencies (such as those with malignancies or acquired immunodeficiency), individuals on immunosuppressive therapy (including corticosteroids and transplant recipients), and people with chronic pulmonary diseases (bronchitis, emphysema, asthma, bronchiectasis, and alveolar proteinosis) . The pathogen is clinically significant due to its ability to disseminate to multiple organ systems, particularly the central nervous system, and its characteristic resistance to multiple antibiotics .
Nocardia farcinica infects host cells through a complex process facilitated by cell wall proteins that enable bacterial adherence to and invasion of host cells. Research has demonstrated that specific virulence factors, such as the Nfa34810 protein, play crucial roles in this process . Nfa34810 has been identified as an immunodominant protein located in the cell wall that facilitates the uptake and internalization of the bacterium into epithelial cells . Experimental evidence shows that coating latex beads with Nfa34810 protein enables their internalization into HeLa cells, and deletion of the nfa34810 gene significantly attenuates the ability of N. farcinica to infect both HeLa and A549 cells .
Accurate identification of Nocardia farcinica in clinical samples involves several methods:
Growth characteristics: N. farcinica can grow at 45°C and shows distinctive opacification when cultured on Middlebrook agar .
Antibiotic susceptibility patterns: Resistance to erythromycin, cefotaxime, and tobramycin is characteristic of N. farcinica and helps differentiate it from other Nocardia species like N. asteroides .
Molecular techniques: Real-time PCR-based high-resolution melting (HRM) analysis targeting the fusA-tuf intergenic region sequence provides clear differentiation of N. farcinica from other Nocardia species such as N. cyriacigeorgica and N. beijingensis . This method offers high sensitivity (detection limit of 10 fg of purified Nocardia DNA) and specificity with no false positives when tested against non-target clinical samples .
Matrix-assisted laser desorption/ionization-time of flight mass spectrometry (MALDI-TOF MS) is also used, though the HRM assay may provide improved identification for certain Nocardia species .
While the search results don't specifically address Pup-protein ligase (pafA), research on other bacterial protein ligases suggests that pafA likely functions in the pupylation pathway, a post-translational modification system analogous to eukaryotic ubiquitination. In mycobacteria and related actinomycetes like Nocardia, pafA catalyzes the ATP-dependent conjugation of the small protein Pup (prokaryotic ubiquitin-like protein) to target proteins, marking them for degradation by the bacterial proteasome. This process is important for bacterial survival under stress conditions and potentially contributes to virulence by helping the bacterium evade host immune responses .
Nfa34810 protein modulates host immune responses through multiple signaling pathways. Research demonstrates that stimulation with Nfa34810 triggers macrophages to produce tumor necrosis factor alpha (TNF-α) through the activation of specific signaling cascades . Specifically, Nfa34810 activates the mitogen-activated protein kinase (MAPK) and nuclear factor κB (NF-κB) signaling pathways by inducing the phosphorylation of several key components:
ERK1/2 (Extracellular signal-regulated kinases 1/2)
p38 (p38 mitogen-activated protein kinases)
JNK (c-Jun N-terminal kinases)
p65 (a component of NF-κB)
Experimental evidence using specific inhibitors of ERK1/2, JNK, and NF-κB shows significantly reduced expression of TNF-α, confirming that Nfa34810-mediated TNF-α production depends on the activation of these kinases . Furthermore, studies using neutralizing antibodies against Toll-like receptor 4 (TLR4) demonstrate significant inhibition of TNF-α secretion, indicating that the Nfa34810-induced inflammatory response operates primarily through the TLR4 pathway .
For studying protein-protein interactions between Nocardia virulence factors (such as pafA or Nfa34810) and host cell receptors, several methodological approaches are recommended:
Co-immunoprecipitation (Co-IP): This technique can identify protein complexes by using antibodies specific to either the bacterial protein or host receptor. For example, anti-TLR4 antibodies could be used to investigate interactions with Nfa34810 .
Surface Plasmon Resonance (SPR): This label-free biophysical technique measures binding affinity and kinetics between purified recombinant bacterial proteins and host receptors.
Yeast Two-Hybrid (Y2H) Screening: While this approach can generate false positives, it offers a high-throughput method to screen for potential interactions.
CRISPR-Cas9 Knockout Studies: Creating knockout cell lines lacking specific receptors (e.g., TLR4) can validate the role of these receptors in recognizing bacterial proteins.
Protein Domain Mapping: Using truncated versions of bacterial proteins to determine which domains are essential for host receptor binding.
Fluorescence Microscopy with Tagged Proteins: This approach can visualize the co-localization of bacterial proteins with host receptors during infection.
Neutralizing Antibody Experiments: Similar to those performed with anti-TLR4 antibodies in Nfa34810 studies, these can block specific receptors to assess their role in bacterial protein recognition .
Expressing and purifying recombinant Nocardia farcinica proteins presents several challenges that researchers must address:
Codon Usage Bias: Nocardia has a high G+C content genome, which can result in codon usage that differs from common expression hosts like E. coli, potentially leading to poor expression levels.
Protein Solubility Issues: Many bacterial cell wall proteins, including those from Nocardia, contain hydrophobic domains that can cause aggregation and inclusion body formation when expressed in heterologous systems.
Post-translational Modifications: If the native protein undergoes specific modifications in Nocardia, these may be absent in recombinant expression systems, potentially affecting protein function.
Protein Toxicity: Some virulence factors may be toxic to expression host cells, limiting yield and requiring tightly regulated expression systems.
Protein Authentication: Confirming that the recombinant protein accurately represents the native protein is crucial. This includes verifying proper folding, oligomerization state, and biological activity.
Endotoxin Contamination: When studying immunomodulatory proteins like Nfa34810, eliminating endotoxin contamination is essential to avoid experimental artifacts in immunological assays .
Assessing the contribution of specific proteins (like pafA or Nfa34810) to Nocardia farcinica virulence in vivo requires several methodological approaches:
Gene Knockout Studies: Creating deletion mutants (like the Δnfa34810 strain) allows direct comparison of virulence between wild-type and mutant bacteria in animal models .
Complementation Studies: Reintroducing the deleted gene on a plasmid or in a different genomic location to confirm that observed attenuated virulence is specifically due to the absence of the target protein.
Animal Infection Models: Using appropriate animal models that recapitulate human disease aspects. For Nocardia infections, both pulmonary and disseminated infection models may be relevant.
Bacterial Burden Quantification: Measuring bacterial loads in various organs to assess the ability of mutant strains to proliferate and disseminate compared to wild-type.
Histopathological Analysis: Examining tissue sections to evaluate differences in inflammatory responses and tissue damage.
Cytokine Profiling: Measuring levels of inflammatory mediators (like TNF-α) in infected tissues or serum to assess immunomodulatory effects .
Survival Studies: Comparing mortality rates between animals infected with wild-type versus mutant bacteria.
Ex vivo Cell Infection Experiments: Using primary cells isolated from animals to bridge in vitro and in vivo studies.
Nocardia farcinica infection involves several key signaling pathways that can be experimentally targeted to understand pathogenesis and potentially develop therapeutic interventions:
| Signaling Pathway | Components Activated | Experimental Targeting Methods | Biological Outcome |
|---|---|---|---|
| MAPK | ERK1/2, p38, JNK | Specific inhibitors (U0126, SB203580, SP600125) | Reduced TNF-α production |
| NF-κB | p65, IκB | Inhibitors (BAY 11-7082), siRNA knockdown | Decreased inflammatory response |
| AKT/PI3K | AKT | Wortmannin, LY294002 inhibitors | Altered cell survival signals |
| TLR4 | MyD88, TRIF | Neutralizing antibodies, knockout models | Blocked recognition of bacterial components |
Research shows that Nfa34810 protein activates these pathways by inducing phosphorylation of multiple signaling components . Experimental evidence demonstrates that specific inhibitors of ERK1/2, JNK, and NF-κB significantly reduce TNF-α expression, confirming the dependency of Nfa34810-mediated TNF-α production on these kinases . The involvement of TLR4 has been confirmed through experiments with neutralizing antibodies against TLR4, which significantly inhibit TNF-α secretion induced by Nfa34810 .
Generating knockout mutants in Nocardia farcinica, such as the Δnfa34810 strain described in the research, requires specialized protocols due to the challenging nature of genetic manipulation in this organism. Based on successful research approaches, the following methodology is recommended:
Target Gene Selection: Identify the gene of interest (e.g., nfa34810 or pafA) using genome databases and bioinformatic analysis to determine gene boundaries and potential polar effects of deletion.
Construct Design: Create a knockout construct containing:
Homologous flanking regions (1-2 kb) upstream and downstream of the target gene
A selectable marker (typically an antibiotic resistance gene compatible with Nocardia)
Optional: Counter-selectable markers for subsequent marker removal
Transformation Method: Electroporation is typically most effective for Nocardia species. Optimize parameters:
Cell preparation: Growth phase is critical (mid-log usually optimal)
DNA concentration: 1-5 μg of purified construct
Electroporation settings: 2.5 kV, 25 μF, 1000 Ω is a standard starting point
Selection and Verification:
Complementation: Create a complementation strain by reintroducing the wild-type gene on a compatible plasmid or at a neutral site in the chromosome to confirm phenotypic changes are specifically due to the target gene deletion.
Optimizing the expression and purification of recombinant Nocardia proteins requires addressing several key challenges:
Expression System Selection:
E. coli BL21(DE3) is commonly used, but for difficult proteins, consider specialized strains like Rosetta (for rare codons) or SHuffle (for disulfide bonds)
For complex proteins, mycobacterial expression systems may provide better folding environments
Vector Design:
Codon optimization for the expression host
Fusion tags: His6, GST, or MBP tags can improve solubility and facilitate purification
Inducible promoters: IPTG-inducible (T7) or auto-induction systems
Expression Conditions Optimization:
Temperature: Lower temperatures (16-25°C) often improve solubility
Induction: Lower IPTG concentrations (0.1-0.5 mM) for slower, more controlled expression
Media: Rich media (TB, 2xYT) or minimal media depending on experimental needs
Additives: 5-10% glycerol, 0.1-0.5 M NaCl, or 1% glucose may improve solubility
Purification Strategy:
Initial capture: Affinity chromatography (Ni-NTA for His-tagged proteins)
Intermediate purification: Ion exchange chromatography
Polishing: Size exclusion chromatography
Tag removal: Site-specific proteases (TEV, PreScission, etc.) if tags interfere with function
Protein Quality Assessment:
SDS-PAGE and Western blotting for purity and identity verification
Dynamic light scattering for aggregation analysis
Circular dichroism for secondary structure confirmation
Functional assays specific to the protein (e.g., enzyme activity, binding assays)
Endotoxin Removal:
Several analytical methods have proven effective for studying host-pathogen protein interactions in Nocardia infections:
Microscopy Techniques:
Confocal microscopy with fluorescently labeled proteins to visualize co-localization
Super-resolution microscopy for detailed interaction visualization
Electron microscopy to examine ultrastructural changes during infection
Biochemical Interaction Analysis:
Co-immunoprecipitation to identify protein complexes
Pull-down assays using purified recombinant proteins
Surface plasmon resonance (SPR) for quantitative binding kinetics
Isothermal titration calorimetry (ITC) for thermodynamic parameters
Proteomic Approaches:
Mass spectrometry-based interaction proteomics
Crosslinking mass spectrometry to capture transient interactions
SILAC or TMT labeling for quantitative interaction differences
Functional Validation Methods:
Structural Biology Approaches:
X-ray crystallography of protein complexes
Cryo-EM for larger assemblies
NMR for studying dynamic interactions
Reporter Systems:
Luciferase reporter assays for signaling pathway activation
FRET/BRET-based interaction detection
Split-GFP complementation for validating interactions in living cells
Designing experiments to differentiate between direct and indirect effects of bacterial proteins (like pafA or Nfa34810) on host signaling pathways requires careful methodological approaches:
Temporal Analysis:
Time-course experiments to determine the sequence of signaling events
Rapid signaling responses (within minutes) often suggest direct effects
Delayed responses may indicate secondary signaling cascades
Dose-Response Relationships:
Titration of purified bacterial protein to establish concentration-dependent effects
Saturation kinetics analysis to determine if responses follow receptor-ligand binding models
In Vitro Reconstitution:
Cell-free systems with purified components to test direct interactions
Kinase assays with purified signaling proteins and bacterial factors
Receptor Blocking/Depletion:
Signaling Pathway Inhibition:
Specific inhibitors targeting different levels of signaling pathways
Compare effects of upstream versus downstream inhibitors
Protein Modification/Truncation:
Structure-function analysis using truncated or mutated bacterial proteins
Identification of specific domains responsible for host interactions
Direct Binding Assays:
Surface plasmon resonance (SPR) or biolayer interferometry
Fluorescence polarization with labeled proteins
Microscale thermophoresis for detecting direct interactions
Genetic Approaches:
Epistasis analysis using combinations of bacterial mutants and host cell pathway inhibitors
Rescue experiments by expressing specific pathway components
Comparing virulence mechanisms between Nocardia farcinica and other pathogenic actinomycetes reveals important evolutionary and functional insights:
Mycobacterium tuberculosis vs. Nocardia farcinica:
Both employ cell wall components that modulate host immune responses
While M. tuberculosis primarily employs the ESX secretion systems for virulence factor delivery, Nocardia appears to rely more on cell wall-associated proteins like Nfa34810
Both activate TLR pathways, though with different receptor specificities (TLR2/4)
Both can survive within macrophages, though through potentially different mechanisms
Rhodococcus equi vs. Nocardia farcinica:
Both are opportunistic pathogens with environmental reservoirs
R. equi virulence is plasmid-associated, while Nocardia virulence factors are chromosomally encoded
Both modulate macrophage activation, though through distinct signaling pathways
Corynebacterium diphtheriae vs. Nocardia farcinica:
C. diphtheriae relies heavily on toxin production, while Nocardia appears to use multiple virulence factors
Nocardia shows broader tissue tropism compared to C. diphtheriae
Different patterns of immune evasion and intracellular survival
Common Features Across Pathogenic Actinomycetes:
Advanced genomic and proteomic approaches offer powerful tools for understanding Nocardia farcinica virulence factors:
Comparative Genomics:
Whole genome sequencing of clinical isolates with varying virulence
Identification of genomic islands associated with pathogenicity
Comparative analysis with non-pathogenic Nocardia species to identify virulence-associated genes
Phylogenetic analysis to understand the evolution of virulence mechanisms
Transcriptomics:
RNA-Seq analysis of N. farcinica during infection of different host cell types
Dual RNA-Seq to simultaneously analyze bacterial and host gene expression
Identification of virulence factor expression patterns under different stress conditions
Small RNA profiling to identify regulatory RNAs involved in virulence
Proteomics:
Quantitative proteomics to identify differentially expressed proteins during infection
Secretome analysis to identify exported virulence factors
Post-translational modification mapping (particularly pupylation catalyzed by pafA)
Protein interaction networks to understand virulence factor connectivity
Functional Genomics:
Transposon mutagenesis with deep sequencing (Tn-Seq) to identify essential genes
CRISPR interference screens to systematically assess gene function
Conditional gene expression systems to study essential virulence factors
Structural Genomics/Proteomics:
High-throughput structural determination of virulence factors
Structure-based drug design targeting key virulence proteins
Computational modeling of protein-protein interactions
The development of targeted therapeutics against Nocardia farcinica virulence factors offers promising approaches for treatment, particularly given the challenges of antibiotic resistance:
Antivirulence Strategies:
Small molecule inhibitors targeting key virulence factors (e.g., Nfa34810, pafA)
Peptide-based inhibitors designed to block protein-protein interactions
Disruption of bacterial signaling systems that regulate virulence factor expression
Immunomodulatory Approaches:
Combination Therapies:
Antivirulence compounds combined with conventional antibiotics
Targeting multiple virulence pathways simultaneously to reduce resistance development
Host-directed therapies combined with bacterial-targeted approaches
Current Drug Susceptibility Patterns:
| Antibiotic | Susceptibility |
|---|---|
| Amikacin | Sensitive |
| Sulfamethoxazole | Sensitive |
| Amoxicillin/clavulanate | Sensitive |
| Imipenem | Sensitive |
| Linezolid | Sensitive |
| Kanamycin | RESISTANT |
| Tobramycin | RESISTANT |
| Gentamicin | RESISTANT |
| Doxycycline | RESISTANT |
| Cefotaxime | RESISTANT |
| Ciprofloxacin | RESISTANT |
| Ceftriaxone | RESISTANT |
| Clarithromycin | RESISTANT |
Therapeutic Development Challenges:
Limited animal models for nocardiosis
Difficulty accessing intracellular bacteria
Need for extended treatment periods
Potential toxicity of new compounds
Accurately differentiating between Nocardia species requires a combination of traditional and molecular approaches:
Phenotypic Methods:
Molecular Identification Techniques:
16S rRNA gene sequencing: The gold standard but may not distinguish closely related species
Multi-locus sequence typing (MLST): Using multiple housekeeping genes for better resolution
Real-time PCR with high-resolution melting (HRM) analysis: Targeting the fusA-tuf intergenic region for rapid, sensitive (10 fg detection limit) and specific differentiation between N. farcinica, N. cyriacigeorgica, and N. beijingensis
Whole genome sequencing: The most comprehensive but resource-intensive approach
Protein-Based Methods:
Combined Approach Algorithm:
Studying host-pathogen interactions in Nocardia infections requires specialized approaches due to the unique characteristics of these bacteria:
Cellular Infection Models:
Selection of appropriate cell lines: A549 (alveolar epithelial), HeLa, and macrophage cell lines (THP-1, RAW264.7) are commonly used
Primary cells: Alveolar macrophages or peripheral blood monocyte-derived macrophages provide more physiologically relevant models
3D cell culture systems: Organoids or air-liquid interface cultures for respiratory tract infections
Infection Parameters Optimization:
Bacterial preparation: Single-cell suspensions (avoiding clumps)
Multiplicity of infection (MOI): Typically 10:1 to 50:1 (bacteria:cells)
Time points: Both early (1-4 hours) and late (24-72 hours) to capture different interaction phases
Readout Systems:
Microscopy: Fluorescence microscopy using GFP-expressing bacteria or immunofluorescence
Flow cytometry: Quantifying infection rates and host cell responses
Cytokine profiling: ELISA or multiplex assays for inflammatory mediators like TNF-α
Gene expression analysis: qRT-PCR or RNA-Seq for host response genes
Signaling Pathway Analysis:
Receptor Studies: