The recombinant Legionella pneumophila subsp. pneumophila UPF0391 membrane protein lpg2415 (UniProt ID: Q5ZSV1) is a bioengineered version of a naturally occurring bacterial membrane protein. It is expressed in E. coli and contains an N-terminal His-tag for purification and detection. Key features include:
Gene Name: lpg2415
Protein Length: Full-length (1–63 amino acids)
AA Sequence: MLYWALIFLIVAIVAGLFGFRGVASAATGIAKVLFFLFIVMFIVLLVFSLLGGTPEPVVI VKP
Storage Buffer: Tris/PBS-based buffer with 6% trehalose (pH 8.0)
Membrane Integration: lpg2415 is classified as an integral membrane protein, with hydrophobic regions suggesting transmembrane domains. Its C-terminal region (residues 44–63) includes hydrophobic stretches, critical for membrane anchoring .
DUF1328 Domain: While not explicitly confirmed in the provided sources, DUF1328 (Domain of Unknown Function 1328) is often linked to membrane proteins with roles in bacterial stress responses or host interaction .
While lpg2415’s precise function remains uncharacterized, its classification as a UPF0391 family protein suggests potential involvement in:
ELISA Assays: Recombinant lpg2415 serves as an antigen for detecting anti-Legionella antibodies in clinical or research settings .
Structural Studies: His-tagged variants enable crystallization or NMR studies to elucidate membrane protein folding .
Functional Characterization: No studies explicitly link lpg2415 to Legionella virulence or metabolic processes. Transposon mutagenesis screens (e.g., InSeq) could clarify its role in mammalian or amoebal hosts .
Structural Analysis: High-resolution studies (e.g., cryo-EM) are needed to map its membrane topology and potential ligand-binding sites.
KEGG: lpn:lpg2415
STRING: 272624.lpg2415
The lpg2415 gene encodes a UPF0391 membrane protein in Legionella pneumophila subspecies pneumophila, specifically characterized in the Philadelphia 1 strain (ATCC 33152/DSM 7513). This protein belongs to the UPF0391 family of uncharacterized membrane proteins. The gene is located within the L. pneumophila genome as part of its core genome, though its genomic neighborhood may vary between different strains. The protein consists of 63 amino acids with the sequence mLYWALIFLIVAIVAGLFGFRGVASAATGIAKVLFFLFIVMFIVLLVFSLLGGTPEPVVI VKP, suggesting a hydrophobic membrane-associated structure with potential transmembrane domains.
While lpg2415 belongs to the UPF0391 family of membrane proteins, comparative genomic analyses have not extensively characterized its homologs across bacterial species. Researchers investigating this protein should perform comparative sequence analyses using tools such as BLAST against other bacterial proteomes to identify potential homologs. The protein may represent a specialized adaptation in Legionella, potentially related to its unique lifestyle as an intracellular pathogen capable of replicating within diverse protozoan hosts and human macrophages. Given that L. pneumophila has acquired numerous genes enabling its pathogenicity, understanding the evolutionary context of lpg2415 through phylogenetic analysis would provide valuable insights into its functional significance.
For successful expression of recombinant lpg2415, researchers should consider several key parameters given its small size and membrane-associated nature. Expression systems using E. coli BL21(DE3) with specialized vectors containing solubility-enhancing fusion tags (such as SUMO, MBP, or TRX) can improve yield and solubility. For membrane proteins like lpg2415, expression at lower temperatures (16-20°C) after induction with reduced IPTG concentrations (0.1-0.5 mM) typically results in better folding and less aggregation. Given the hydrophobic nature of lpg2415, specialized membrane protein expression systems or cell-free expression systems may provide advantages. If faced with toxicity issues, consider using tightly regulated expression systems or C41/C45 E. coli strains specifically designed for toxic membrane proteins.
Purification of lpg2415 requires specialized approaches due to its membrane-associated nature. The recommended workflow includes:
Membrane fraction isolation using ultracentrifugation (100,000 × g) after cell lysis
Solubilization with appropriate detergents (e.g., n-dodecyl-β-D-maltopyranoside (DDM), n-octyl-β-D-glucopyranoside (OG), or lauryl maltose neopentyl glycol (LMNG))
Affinity chromatography using the appropriate tag system
Size exclusion chromatography for final purification
For storage, the purified protein should be maintained in a Tris-based buffer with 50% glycerol at -20°C or -80°C, avoiding repeated freeze-thaw cycles. For short-term work, aliquots can be stored at 4°C for up to one week. When designing purification protocols, researchers should monitor protein stability at each step using dynamic light scattering or thermal shift assays to ensure the membrane protein remains properly folded.
Assessing the functional activity of lpg2415 presents challenges due to its uncharacterized nature. Several approaches can be employed:
Lipid binding assays using fluorescence anisotropy or surface plasmon resonance to detect interactions with different membrane components
Reconstitution into liposomes or nanodiscs followed by biophysical characterization
Protein-protein interaction studies using pull-down assays, co-immunoprecipitation, or yeast two-hybrid systems to identify binding partners within L. pneumophila
Electrophysiological methods if the protein functions as an ion channel or transporter
Researchers should also consider functional complementation experiments in lpg2415 knockout strains to observe phenotypic changes related to virulence, stress response, or intracellular survival. Given L. pneumophila's complex pathogenic mechanisms, investigating lpg2415's potential interactions with host factors during infection could reveal insights into its functional role.
L. pneumophila strains exhibit varying levels of resistance to complement-mediated killing, a phenotype associated with enhanced virulence. Recent population genomic studies have identified specific genes strongly associated with this resistance, particularly lag-1, which encodes an O-acetyltransferase responsible for lipopolysaccharide modification. While the direct relationship between lpg2415 and complement resistance has not been definitively established, membrane proteins can contribute to complement evasion mechanisms. Researchers should investigate potential associations through:
Comparative genomic analyses of clinical versus environmental isolates to determine if lpg2415 variants correlate with virulence
Serum resistance assays comparing wild-type and lpg2415 mutant strains
Analysis of lpg2415's potential interaction with outer membrane components involved in complement resistance
Population genomic studies have revealed that certain L. pneumophila sequence types are responsible for a disproportionate number of human infections, suggesting specific genetic factors contribute to enhanced pathogenicity. Investigating whether lpg2415 is conserved or variable across these lineages could provide insights into its potential role in virulence.
Understanding the expression pattern of lpg2415 during L. pneumophila's life cycle could provide insights into its function. L. pneumophila undergoes significant physiological changes as it transitions between environmental survival, intracellular replication, and transmission phases. Researchers should examine lpg2415 expression under various conditions using:
Quantitative RT-PCR analysis comparing expression across growth phases (exponential, stationary) and environmental conditions (temperature, pH, nutrient limitation)
RNA-seq analysis of bacteria during different stages of intracellular infection
Reporter gene constructs (e.g., lpg2415 promoter fused to GFP) to visualize expression dynamics
Proteomic approaches to confirm protein-level expression
L. pneumophila employs sophisticated regulatory networks to control virulence factor expression, including the Legionella-containing vacuole formation required for intracellular replication. Determining whether lpg2415 is regulated by known virulence-associated transcription factors would provide evidence for its potential role in pathogenesis.
Identifying protein interaction partners is crucial for understanding lpg2415's function. Advanced techniques for mapping protein-protein interactions include:
Bacterial two-hybrid systems optimized for membrane proteins
Proximity labeling approaches (BioID, APEX) to identify proteins in close proximity to lpg2415 within native cellular contexts
Chemical crosslinking combined with mass spectrometry (XL-MS) to capture transient interactions
Co-immunoprecipitation with anti-lpg2415 antibodies followed by mass spectrometry
Blue native PAGE to preserve membrane protein complexes for analysis
These approaches should be applied to both laboratory cultures and bacteria recovered from infection models to identify condition-specific interactions. The type IVB secretion system is crucial for L. pneumophila pathogenesis, and determining whether lpg2415 interacts with components of this system would be particularly valuable. Additionally, constructing interaction networks that place lpg2415 within the context of known virulence pathways could reveal its functional role.
CRISPR-Cas genome editing offers powerful approaches for functional characterization of lpg2415:
Generation of precise deletion or point mutations in lpg2415 to assess phenotypic consequences
CRISPRi (interference) for conditional knockdown of lpg2415 expression
CRISPRa (activation) to upregulate lpg2415 expression
CRISPR-mediated base editing for introducing specific amino acid substitutions
Pooled CRISPR screens in infection models to assess the contribution of lpg2415 to fitness during infection
When designing CRISPR-Cas experiments in L. pneumophila, researchers should optimize protocols for transformation efficiency and consider the bacterium's complex genome with potential redundancy in virulence functions. Phenotypic assessment should include growth kinetics, stress resistance, intracellular survival in amoebae and macrophages, and virulence in animal models.
Advanced computational methods can provide functional predictions for uncharacterized proteins like lpg2415:
Structural prediction using AlphaFold2 or RoseTTAFold to generate high-confidence 3D models
Molecular dynamics simulations to analyze membrane integration and conformational dynamics
Comparative genomic analyses across Legionella species and strains to identify conservation patterns and genetic linkage
Analysis of genomic island localization to determine if lpg2415 was acquired through horizontal gene transfer
Co-expression network analysis to identify functionally related genes
Recent genomic characterization studies of Legionella species have identified conserved and variable components across the genus. Determining whether lpg2415 belongs to the core genome or accessory genome of Legionella would provide evolutionary context for its function. Additionally, examining synteny and genetic linkage with known virulence factors could suggest functional associations.
Population genomic studies have revealed significant differences between L. pneumophila strains associated with human disease and those found predominantly in environmental samples. Researchers investigating lpg2415's potential role in virulence should:
Compare the presence, sequence variation, and expression levels of lpg2415 across large collections of clinical and environmental isolates
Perform genome-wide association studies (GWAS) to determine if specific lpg2415 variants associate with clinical origin
Analyze lpg2415 in the context of dominant sequence types responsible for human infections
Examine lpg2415 conservation across different L. pneumophila serogroups
Research has shown that a limited number of L. pneumophila sequence types are responsible for almost half of all human infections, suggesting specific genetic factors contribute to enhanced human pathogenicity. Determining whether lpg2415 is uniquely conserved or exhibits specific variants in these lineages would provide evidence for its potential role in human disease.
Developing specific antibodies against lpg2415 provides valuable tools for research and potentially for diagnostics:
Immunolocalization studies to determine the precise cellular localization of lpg2415
Western blot analysis to monitor expression levels under different conditions
Flow cytometry to analyze surface exposure on intact bacteria
Immunoprecipitation for protein interaction studies
Potential diagnostic applications if lpg2415 proves to be accessible on the bacterial surface
When developing antibodies against small membrane proteins like lpg2415, researchers should carefully select antigenic peptide sequences, preferably from predicted exposed regions. Consideration should be given to both polyclonal antibodies for maximum epitope coverage and monoclonal antibodies for specificity in complex samples. Validation should include specificity testing against lpg2415 knockout strains.
Evaluating lpg2415 as a potential therapeutic target requires thorough characterization of its role in bacterial survival and pathogenesis:
Essentiality screening to determine if lpg2415 is required for bacterial viability
Assessment of lpg2415's contribution to antibiotic resistance mechanisms
In vitro and in vivo infection models comparing wild-type and lpg2415 mutant strains
High-throughput screening for small molecule inhibitors if function is established
Evaluation of conservation across clinically relevant strains to assess target breadth
L. pneumophila contains virulence factors that have been distributed horizontally across major phylogenetic clades, suggesting selective pressure for their maintenance. If lpg2415 proves to be widely conserved and involved in virulence, it could represent a valuable target for novel therapeutic approaches. Importantly, membrane proteins often make attractive drug targets due to their accessibility and essential functions.