KEGG: hpj:jhp_0078
STRING: 85963.jhp0078
Helicobacter pylori is a Gram-negative, helix-shaped, microaerophilic bacterium belonging to the family Helicobacteraceae. It was identified in 1982 by Australian scientists Barry Marshall and Robin Warren. This pathogen colonizes the upper gastrointestinal tract in more than 50% of the global population. Its clinical significance stems from its strong association with serious gastrointestinal conditions - infected individuals face a 10-20% lifetime risk of developing peptic ulcers and a 1-2% risk of acquiring stomach cancer. H. pylori has also been linked to colorectal polyps and colorectal cancer development, making it a critical target for medical research focused on gastrointestinal diseases .
The jhp_0078 protein is an uncharacterized protein from Helicobacter pylori. The recombinant form that has been studied includes amino acids 1-62 of the native protein. While detailed structural and functional characterization remains limited in current literature, this protein represents one of the many expressed proteins in H. pylori that may contribute to its pathogenicity and survival mechanisms. As with many bacterial proteins, understanding its structure and function could provide insights into bacterial pathogenesis and potential therapeutic targets .
Studying uncharacterized proteins like jhp_0078 is essential because H. pylori is highly polymorphic, with certain strains demonstrating significantly higher disease-causing potential than others. Uncharacterized proteins may play crucial roles in virulence, antibiotic resistance, immune evasion, or biofilm formation. By systematically investigating these proteins, researchers can identify novel virulence factors, potential drug targets, or biomarkers for disease severity. The methodological approach involves comparative genomics, structural analysis, and functional studies to determine the role of these proteins in bacterial pathogenesis and their potential as therapeutic targets .
For recombinant jhp_0078 production, researchers have several expression system options, each with specific advantages depending on research goals. The protein can be expressed in E. coli, yeast, baculovirus, or mammalian cell systems. The methodological approach should begin with codon optimization based on the chosen expression system. For prokaryotic expression in E. coli, researchers should consider factors such as protein hydrophilicity, codon rarity, and potential protein toxicity that may affect expression efficiency. For eukaryotic expression in yeast, baculovirus, or mammalian cells, post-translational modifications may be better preserved. The selection should be guided by downstream applications - if structural studies are planned, prokaryotic systems may be sufficient, while functional studies might benefit from eukaryotic expression systems that maintain native protein folding and modifications .
When expressing full-length jhp_0078, researchers may encounter truncated products due to proteolysis or improper translation initiation. To address this methodological challenge, implement a dual-tagged expression strategy using fusion tags at both N and C termini (such as His-tag and GST or FLAG). This approach allows for differentiation between full-length proteins and truncated products during purification. Additionally, optimize elution conditions by using an increasing imidazole concentration gradient during purification to separate full-length proteins from truncated variants. For expression vector design, include strong ribosome binding sites and optimal spacing between the promoter and start codon. Consider codon optimization of the 5' region to enhance translation initiation efficiency. Finally, test multiple expression conditions (temperature, induction time, inducer concentration) to minimize proteolytic degradation during expression .
Recombinant jhp_0078 has several research applications in the study of H. pylori pathogenesis. Methodologically, the protein can be utilized in structural biology studies to determine its three-dimensional structure using X-ray crystallography or cryo-electron microscopy, providing insights into its potential function. In immunological studies, the recombinant protein can serve as an antigen for developing antibodies that enable detection and localization studies within bacterial cells or during host infection. For functional characterization, protein-protein interaction studies using techniques like pull-down assays, co-immunoprecipitation, or yeast two-hybrid systems can identify binding partners and potential roles in bacterial physiology. The protein may also be valuable in vaccine development research, where it could be assessed for immunogenicity and protective efficacy in animal models. Additionally, the recombinant protein enables epitope mapping studies to identify regions recognized by the host immune system during infection .
The relationship between biofilm formation and jhp_0078 expression requires sophisticated methodological approaches. Biofilm formation in H. pylori serves as a survival mechanism against antibiotics, immune attack, and environmental stresses, potentially altering the expression profile of proteins including jhp_0078. To investigate this relationship, researchers should employ a comparative transcriptomics and proteomics approach between planktonic and biofilm-growing H. pylori isolates. Methodologically, biofilm formation can be quantified using crystal violet assays on glass coverslips, while protein expression can be measured using RNA-seq and quantitative proteomics. Functional studies should then examine whether jhp_0078 contributes to biofilm formation through knockout/knockdown studies and complementation experiments. Microscopy techniques including confocal microscopy with fluorescently-tagged jhp_0078 can determine its localization within biofilm structures. Such studies are critical since biofilm-associated proteins may represent targets for disrupting bacterial persistence during chronic infection .
When investigating protein-protein interactions involving jhp_0078, a rigorously designed experimental approach is essential. The methodological framework should begin with preliminary bioinformatic analysis using tools like STRING, IntAct, or BIOGRID to predict potential interaction partners based on co-expression, genomic context, or homology to known interaction networks. For experimental validation, employ multiple complementary techniques: (1) Co-immunoprecipitation with antibodies against jhp_0078 followed by mass spectrometry identification of binding partners, similar to approaches used in CD40-TSHR interaction studies; (2) Proximity labeling methods such as BioID or APEX2 to identify proteins in close proximity to jhp_0078 in vivo; (3) Yeast two-hybrid or bacterial two-hybrid screening to identify direct protein-protein interactions; and (4) Surface plasmon resonance or isothermal titration calorimetry to quantify binding affinities and kinetics. Control experiments must include negative controls (unrelated proteins), validation with reverse pull-downs, and verification in different experimental conditions. This multi-technique approach minimizes false positives and provides confidence in identified interaction partners .
To determine jhp_0078's potential role in H. pylori virulence, researchers should implement a multi-faceted genomic approach. Methodologically, begin with comparative genomics by sequencing multiple clinical isolates from patients with varying disease severity (from asymptomatic to severe gastric cancer) using hybrid assembly of Nanopore MinION and Illumina MiSeq data. This enables identification of polymorphisms or expression variations in jhp_0078 that correlate with disease outcomes. Create isogenic mutant strains using homologous recombination or CRISPR-Cas9 technology to delete or modify jhp_0078, then assess virulence phenotypes in cell culture infection models and animal models. Measure epithelial cell adhesion, invasion, inflammatory cytokine induction, and gastric colonization efficiency. Transcriptomic analysis using RNA-seq comparing wild-type and mutant strains under various growth conditions (pH stress, antibiotic exposure, host cell contact) can reveal co-regulated genes and potential virulence-associated pathways involving jhp_0078. This comprehensive approach provides mechanistic insights into jhp_0078's contribution to bacterial pathogenesis .
Studying post-translational modifications (PTMs) of jhp_0078 requires sophisticated analytical approaches. The methodological workflow should begin with computational prediction of potential PTM sites using algorithms specific for phosphorylation (NetPhos), glycosylation (NetNGlyc, NetOGlyc), acetylation (PAIL), and other modifications. For experimental verification, express and purify recombinant jhp_0078 from both prokaryotic (E. coli) and eukaryotic (mammalian or insect) expression systems to compare modification patterns. Employ high-resolution mass spectrometry techniques including LC-MS/MS with electron transfer dissociation (ETD) or higher-energy collisional dissociation (HCD) fragmentation to precisely map modification sites. Site-directed mutagenesis of predicted PTM sites followed by functional assays can determine the biological significance of identified modifications. For in vivo confirmation, develop modification-specific antibodies or use enrichment strategies (e.g., titanium dioxide for phosphopeptides, lectin affinity for glycopeptides) prior to mass spectrometry analysis. Additionally, investigate changes in PTM patterns under different growth conditions or during host cell interaction to understand their regulatory roles in pathogenesis .
Optimizing expression conditions for recombinant jhp_0078 requires systematic methodological refinement across multiple parameters. Begin with expression vector selection, comparing constructs with different promoters (T7, tac, or pBAD), fusion tags (His, GST, MBP, or SUMO), and codon optimization for the expression host. For E. coli expression, test multiple strains including BL21(DE3), Rosetta(DE3), or Arctic Express to address potential codon bias or protein folding issues. Conduct a temperature optimization matrix (15°C, 20°C, 25°C, 30°C, and 37°C) with varied IPTG concentrations (0.1mM to 1mM) and induction durations (3h, 6h, overnight, 24h). For purification, implement a multi-step approach beginning with affinity chromatography (Ni-NTA for His-tagged constructs), followed by ion exchange chromatography and size exclusion chromatography to achieve >95% purity. Protein quality should be assessed through SDS-PAGE, Western blotting, mass spectrometry, and circular dichroism to verify structural integrity. For problematic expression, consider alternative approaches such as cell-free protein synthesis systems or secretion-based expression strategies to reduce toxicity or aggregation issues. Document yield at each optimization step to identify critical parameters affecting expression efficiency .
Designing experiments to assess jhp_0078 immunogenicity requires a comprehensive methodological framework spanning in silico prediction to in vivo validation. Begin with computational epitope prediction using algorithms like BepiPred, DiscoTope, or IEDB tools to identify potential B-cell and T-cell epitopes within jhp_0078. Express and purify recombinant jhp_0078 with confirmed structural integrity through circular dichroism or thermal shift assays. For in vitro assessment, conduct T-cell proliferation assays using peripheral blood mononuclear cells (PBMCs) from H. pylori-infected and uninfected individuals stimulated with purified jhp_0078, measuring proliferation by CFSE dilution or thymidine incorporation. Characterize the cytokine profile (IFN-γ, IL-4, IL-17) by ELISA or intracellular cytokine staining to determine T-helper polarization. For B-cell responses, measure jhp_0078-specific antibodies in patient sera using ELISA, and assess functional activity through neutralization or opsonization assays. In animal models, immunize mice with recombinant jhp_0078 using various adjuvants, then challenge with H. pylori to evaluate protective efficacy by measuring bacterial colonization, histopathological changes, and immune cell infiltration. This systematic approach provides comprehensive insights into jhp_0078's potential as a vaccine candidate or diagnostic marker .
Comprehensive structural characterization of jhp_0078 requires a multi-technique methodological approach to analyze different aspects of protein structure. For primary structure confirmation, employ high-resolution mass spectrometry (MS) techniques including LC-MS/MS peptide mapping to verify the amino acid sequence and identify any post-translational modifications. Secondary structure analysis should utilize circular dichroism (CD) spectroscopy to quantify α-helical, β-sheet, and random coil content, complemented by Fourier-transform infrared spectroscopy (FTIR) to confirm secondary structural elements. For tertiary structure determination, use X-ray crystallography after optimizing crystallization conditions through systematic screening of precipitants, buffers, and additives. If crystallization proves challenging, employ nuclear magnetic resonance (NMR) spectroscopy for proteins under 20 kDa or cryo-electron microscopy (cryo-EM) for larger assemblies. Supplement these approaches with small-angle X-ray scattering (SAXS) to obtain low-resolution structural information in solution. For thermal stability assessment, utilize differential scanning calorimetry (DSC) and thermal shift assays to determine melting temperatures under varying pH and buffer conditions. Additionally, apply hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify solvent-accessible regions and potential binding interfaces that may be functionally relevant .
Developing diagnostic approaches using jhp_0078 requires methodological innovation across multiple platforms. Begin with bioinformatic analysis to assess sequence conservation of jhp_0078 across diverse H. pylori strains and its absence in closely related bacterial species to confirm specificity. For serological diagnostics, develop ELISAs using purified recombinant jhp_0078 to detect specific antibodies in patient sera, establishing sensitivity and specificity through ROC curve analysis with samples from confirmed H. pylori-positive and negative individuals. For molecular diagnostics, design and validate jhp_0078-specific PCR primers and probes for conventional, real-time, and multiplex PCR assays to detect H. pylori directly from biopsy specimens or stool samples. Develop rapid point-of-care tests using lateral flow immunoassay technology with anti-jhp_0078 antibodies for antigen detection in clinical samples. For advanced applications, explore aptamer-based biosensors using SELEX technology to select high-affinity aptamers against jhp_0078, which can be incorporated into electrochemical or optical biosensing platforms. Validate all diagnostic approaches through comparative studies with established diagnostic methods (urea breath test, histopathology, culture) using sensitivity, specificity, positive and negative predictive values as performance metrics .
Investigating jhp_0078's potential role in antibiotic resistance requires a methodical research approach. Begin with comparative genomics and transcriptomics analysis of antibiotic-resistant versus susceptible H. pylori strains to determine if jhp_0078 expression correlates with resistance phenotypes. Generate isogenic knockout mutants (Δjhp_0078) and overexpression strains to directly assess changes in minimum inhibitory concentrations (MICs) for various antibiotic classes (clarithromycin, metronidazole, amoxicillin, tetracycline, levofloxacin) using standardized broth microdilution or Etest methods. Conduct time-kill assays to evaluate the kinetics of bacterial killing in wild-type versus mutant strains. For mechanistic understanding, perform protein-protein interaction studies using pull-down assays and mass spectrometry to identify if jhp_0078 interacts with known resistance determinants or drug efflux systems. Employ fluorescently labeled antibiotics to track intracellular accumulation in wild-type versus mutant strains using flow cytometry and confocal microscopy. If jhp_0078 affects biofilm formation, assess antibiotic penetration and efficacy within biofilms using confocal microscopy with live/dead staining. Additionally, evaluate stress response activation and morphological changes using transmission electron microscopy to understand physiological adaptations that might contribute to resistance .
Investigating jhp_0078's potential role in H. pylori-associated gastric cancer requires a comprehensive methodological approach spanning epidemiological, molecular, and functional studies. Begin with a case-control epidemiological study comparing jhp_0078 prevalence, sequence variations, and expression levels in H. pylori isolates from patients with gastric cancer versus those with non-malignant conditions. Utilize next-generation sequencing to identify single nucleotide polymorphisms or structural variations in jhp_0078 that may correlate with cancer risk. For in vitro functional studies, expose gastric epithelial cell lines (AGS, MKN45) to purified recombinant jhp_0078 and assess carcinogenic potential through proliferation assays (BrdU incorporation, Ki-67 staining), apoptosis resistance (Annexin V/PI staining, caspase activity), DNA damage (γH2AX foci, comet assay), and oncogenic signaling pathway activation (MAPK, Wnt/β-catenin, STAT3) using Western blotting and reporter assays. Develop three-dimensional organoid cultures from primary gastric tissues to evaluate jhp_0078's effects on tissue architecture and differentiation. For in vivo validation, utilize transgenic mouse models expressing jhp_0078 in the gastric mucosa or infection models with jhp_0078-modified H. pylori strains, monitoring for preneoplastic changes and tumor development through histopathological analysis, immunohistochemistry for cancer markers, and transcriptomic profiling of mucosal tissues .
This methodological comparison table provides researchers with a systematic framework for selecting appropriate experimental approaches when comparing wild-type and mutant jhp_0078 functions. The comprehensive analysis enables identification of structural features critical for function and potential compensatory mechanisms when the protein is altered .
Expression System | Yield Potential | Post-translational Modifications | Expression Time | Cost | Optimal Applications |
---|---|---|---|---|---|
E. coli | High (5-500 mg/L) | Limited (basic folding only) | Fast (1-2 days) | Low | Structural studies; Antibody production; High-throughput screening |
Yeast (P. pastoris, S. cerevisiae) | Medium-High (10-300 mg/L) | Moderate (glycosylation patterns differ from mammalian) | Medium (3-5 days) | Medium | Functional studies requiring some PTMs; Large-scale production |
Baculovirus/Insect cells | Medium (5-50 mg/L) | Good (similar to mammalian except for complex N-glycans) | Slow (7-10 days) | High | Functional studies; Protein-protein interactions; Enzyme activity assays |
Mammalian cells | Low-Medium (1-20 mg/L) | Excellent (native-like glycosylation and other PTMs) | Very slow (2-3 weeks) | Very high | Studies requiring authentic folding and PTMs; Cell-based assays |
Cell-free protein synthesis | Low-Medium (0.5-5 mg/ml reaction) | Limited (depends on lysate source) | Very fast (hours) | Very high | Difficult-to-express proteins; Proteins toxic to cells; Rapid prototyping |
This comparative analysis provides researchers with a methodological framework for selecting the optimal expression system based on their specific research requirements. The decision should be guided by the intended downstream applications, required protein quality, available resources, and time constraints. For initial structural characterization of jhp_0078, E. coli expression may be sufficient, while functional studies examining interactions with host factors would benefit from mammalian or insect cell expression systems that preserve post-translational modifications .
Clinical Outcome | Proposed Experimental Approach | Key Parameters to Measure | Analysis Methods | Expected Correlation if jhp_0078 Contributes to Pathogenesis |
---|---|---|---|---|
Gastritis severity | Clinical sample collection with histological scoring; jhp_0078 detection by PCR and immunohistochemistry | Inflammation score; Neutrophil infiltration; jhp_0078 expression level | Mann-Whitney U test; Spearman correlation; Multivariate regression | Positive correlation between jhp_0078 expression and inflammation scores |
Peptic ulcer development | Case-control study comparing jhp_0078 sequence variants in ulcer vs. non-ulcer patients | jhp_0078 sequence polymorphisms; Gene expression levels; Bacterial load | Chi-square test; Odds ratio calculation; Logistic regression | Higher prevalence of specific jhp_0078 variants in ulcer patients |
Gastric cancer risk | Longitudinal cohort study following H. pylori+ patients; jhp_0078 genotyping at baseline | Cancer development; Precancerous lesions progression; jhp_0078 expression patterns | Kaplan-Meier survival analysis; Cox proportional hazards model | Faster progression to cancer in patients with high-expression jhp_0078 variants |
Treatment resistance | Clinical isolate collection before and after failed treatment; jhp_0078 analysis | Antibiotic MICs; Treatment failure rates; jhp_0078 expression changes | Paired t-test; McNemar's test; Regression analysis | Upregulation of jhp_0078 in post-treatment isolates; Association with MIC values |
Asymptomatic carriage | Community screening with endoscopy follow-up; jhp_0078 characterization | Symptom scores; Endoscopic findings; Bacterial colonization density | ANOVA; Classification algorithms; Clustering analysis | Lower expression or distinct variants in asymptomatic carriers |
This methodological framework provides researchers with a structured approach to investigate potential correlations between jhp_0078 and clinical outcomes in H. pylori infection. The comparative analysis enables identification of jhp_0078 variants or expression patterns that may serve as biomarkers for disease progression or treatment response. Implementation of these approaches requires careful study design, appropriate statistical power calculation, and controlling for confounding variables such as host genetic factors, environmental exposures, and co-infection with other H. pylori virulence factors .
The future research landscape for jhp_0078 should focus on integrating multiple methodological approaches to fully elucidate its biological significance. Structural biology represents a critical frontier, where advanced techniques like AlphaFold2 prediction combined with experimental validation through X-ray crystallography or cryo-EM could reveal functional domains and interaction surfaces. Systems biology approaches using comprehensive protein-protein interaction mapping, transcriptomics, and metabolomics will position jhp_0078 within the broader context of H. pylori's virulence networks. In vivo studies using transgenic mouse models with tissue-specific expression of jhp_0078 could demonstrate its direct effects on gastric pathology independent of other bacterial factors. Single-cell analysis of infected gastric tissues would reveal cell type-specific responses to jhp_0078 exposure. Additionally, evolutionary studies examining jhp_0078 conservation and divergence across H. pylori strains from different geographic regions and disease associations could identify functionally important regions under selective pressure. These multi-disciplinary approaches would collectively enhance our understanding of jhp_0078's role in H. pylori pathogenesis and potentially identify novel therapeutic strategies targeting this protein .
Translating jhp_0078 research into clinical applications requires a systematic methodological pathway from bench to bedside. For diagnostic applications, jhp_0078-specific antibodies or aptamers could be incorporated into rapid point-of-care tests for H. pylori detection, with clinical validation studies measuring sensitivity and specificity against gold standard methods. If jhp_0078 exhibits strain-specific variations associated with virulence, genotyping assays could stratify patients by risk level, enabling personalized treatment decisions. In therapeutic development, if structural and functional studies identify jhp_0078 as essential for bacterial survival or virulence, small molecule inhibitors could be designed through structure-based virtual screening followed by medicinal chemistry optimization. The drug development pathway would include in vitro efficacy testing, pharmacokinetic/pharmacodynamic studies, and eventually clinical trials. For vaccination strategies, if immunogenicity studies confirm jhp_0078 as a protective antigen, vaccine formulations could be developed using recombinant protein, synthetic peptides representing immunodominant epitopes, or DNA/RNA vaccine approaches. Preclinical testing in animal models would assess protective efficacy, followed by Phase I-III clinical trials evaluating safety, immunogenicity, and protection against infection or disease progression. This translational pathway requires collaborative efforts between basic scientists, clinicians, and industry partners to move promising findings toward patient benefit .