Recombinant Uncharacterized SURF1-like protein Rv2235/MT2294 (Rv2235, MT2294)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for fulfillment.
Lead Time
Delivery times vary depending on the purchasing method and location. Please contact your local distributor for precise delivery estimates.
Note: Our proteins are shipped with standard blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquoting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can be used as a reference.
Shelf Life
Shelf life depends on various factors, including storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized forms have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is essential for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-271
Protein Length
full length protein
Target Names
Rv2235, MT2294
Target Protein Sequence
MPRLAFLLRPGWLALALVVVAFTYLCFTVLAPWQLGKNAKTSRENQQIRYSLDTPPVPLK TLLPQQDSSAPDAQWRRVTATGQYLPDVQVLARLRVVEGDQAFEVLAPFVVDGGPTVLVD RGYVRPQVGSHVPPIPRLPVQTVTITARLRDSEPSVAGKDPFVRDGFQQVYSINTGQVAA LTGVQLAGSYLQLIEDQPGGLGVLGVPHLDPGPFLSYGIQWISFGILAPIGLGYFAYAEI RARRREKAGSPPPDKPMTVEQKLADRYGRRR
Uniprot No.

Q&A

What is the genomic context of Rv2235/MT2294 in Mycobacterium tuberculosis?

Rv2235 is situated within an operon structure in the Mycobacterium tuberculosis genome. According to genomic annotation data, the Rv2235 gene exists as part of the Rv2232-Rv2235 operon . Operons in prokaryotic organisms typically contain functionally related genes that are co-transcribed, suggesting that Rv2235 may function in coordination with other proteins coded by genes in this operon.

Understanding the genomic context of Rv2235 requires comprehensive genomic analysis using bioinformatics tools. Researchers should utilize whole-genome sequencing data and comparative genomics approaches to analyze the conservation of this gene across mycobacterial species. The positional relationship between Rv2235 and adjacent genes can provide insights into potential functional relationships and co-expression patterns.

For robust genomic context analysis, researchers should implement both computational prediction methods and experimental verification through techniques such as RT-PCR or RNA-seq to confirm operon structure and co-transcription patterns. This approach helps establish a foundation for understanding the regulatory mechanisms controlling Rv2235 expression and its potential functional partners.

How is recombinant Rv2235/MT2294 protein typically expressed and purified for research?

Expression and purification of recombinant Rv2235/MT2294 protein for research purposes typically follows established protocols for mycobacterial membrane proteins. Based on approaches used for similar proteins, researchers commonly utilize bacterial expression systems such as E. coli BL21(DE3) strains with expression vectors containing affinity tags (His-tag, GST-tag) for simplified purification.

The expression process generally involves cloning the Rv2235 gene into an appropriate expression vector with an inducible promoter system (such as T7 or pET). Expression conditions require optimization, including temperature (often 18-25°C for membrane proteins), inducer concentration, and duration of induction. Due to potential toxicity or inclusion body formation, specialized E. coli strains like C41(DE3) or C43(DE3) designed for membrane protein expression may yield better results.

Purification typically involves initial cell lysis using methods suitable for membrane proteins (e.g., sonication or French press), followed by membrane fraction isolation through ultracentrifugation. The protein can be solubilized using appropriate detergents (e.g., n-dodecyl-β-D-maltoside or CHAPS) before affinity chromatography. Further purification may employ size-exclusion chromatography to obtain homogeneous protein preparations. Throughout the process, protein integrity should be verified using methods like SDS-PAGE and Western blotting with antibodies specific to Rv2235 or the affinity tag.

What basic biochemical assays are used to characterize Rv2235/MT2294 protein function?

Characterization of uncharacterized proteins like Rv2235/MT2294 typically begins with a series of fundamental biochemical assays to establish basic functional parameters. Given its classification as a SURF1-like protein, researchers should investigate potential roles in respiratory function or protein assembly processes.

Initial biochemical characterization should include protein-protein interaction studies using co-immunoprecipitation or pull-down assays to identify binding partners, similar to approaches used in studying other mycobacterial proteins . These techniques can reveal associations with components of respiratory complexes or other cellular machinery. Researchers should also employ enzymatic activity assays based on predicted functions from bioinformatic analyses, testing for activities related to cytochrome c oxidase assembly or other respiratory chain functions.

Additionally, subcellular localization studies using fractionation techniques and immunolocalization can provide insights into where Rv2235 operates within the mycobacterial cell. This information is critical for contextualizing its function. Researchers should complement these approaches with stability assays under various environmental conditions (pH, temperature, oxidative stress) to understand the protein's behavior under conditions relevant to tuberculosis pathogenesis. For more detailed functional insights, site-directed mutagenesis of conserved residues followed by functional assays can identify critical domains essential for protein activity.

How does Rv2235/MT2294 expression correlate with drug resistance in clinical M. tuberculosis isolates?

The correlation between Rv2235/MT2294 expression and drug resistance in clinical M. tuberculosis isolates represents an important research question with potential implications for understanding tuberculosis treatment outcomes. While current research has not specifically focused on Rv2235, methodological approaches similar to those used in studying efflux pump genes can be applied.

Researchers should employ quantitative real-time PCR (RT-qPCR) to measure Rv2235 expression levels across drug-sensitive and drug-resistant clinical isolates, normalizing expression against established housekeeping genes like polA . This approach allows for calculation of relative expression using methods such as the 2−ΔΔCT method, with expression levels above 4-fold commonly considered significant overexpression, similar to thresholds used for established efflux pump genes .

For comprehensive analysis, this expression data should be correlated with minimum inhibitory concentration (MIC) values determined using standardized methods such as the resazurin microtiter assay (REMA) . Researchers should also sequence the Rv2235 gene and its promoter region to identify any mutations that may correlate with altered expression or drug response profiles. Additionally, creating knockout or overexpression strains of Rv2235 in laboratory M. tuberculosis strains would allow direct assessment of its impact on drug susceptibility profiles through comparative MIC determination with and without efflux pump inhibitors like carbonyl cyanide 3-chlorophenylhydrazone (CCCP) .

What structural similarities exist between Rv2235/MT2294 and other characterized SURF1 proteins?

Structural analysis of Rv2235/MT2294 in relation to characterized SURF1 proteins requires sophisticated computational and experimental approaches. SURF1 proteins are known to function in cytochrome c oxidase assembly, with mutations linked to conditions like Leigh syndrome in humans , suggesting potential functional conservation across species.

Researchers should begin with comprehensive sequence alignment of Rv2235 against characterized SURF1 proteins from various organisms, focusing on conserved domains and motifs. Advanced structural prediction tools employing machine learning algorithms can generate tertiary structure models of Rv2235 based on homology to known SURF1 structures. These predictions should be validated through experimental approaches such as X-ray crystallography or cryo-electron microscopy when possible.

Key structural features to investigate include transmembrane domains, substrate binding pockets, and potential interaction interfaces with partner proteins. For more detailed structural analysis, researchers should examine conservation of specific amino acid residues known to be critical for SURF1 function in other organisms, particularly those associated with pathological mutations in human SURF1 . Site-directed mutagenesis of these conserved residues in recombinant Rv2235, followed by functional assays, can provide experimental validation of structural predictions and establish structure-function relationships.

Additionally, molecular dynamics simulations can provide insights into protein flexibility, conformational changes, and potential mechanisms of action. These computational approaches should be integrated with experimental data from techniques like circular dichroism spectroscopy or limited proteolysis to assess secondary structure content and domain organization, respectively.

What methodologies are most effective for investigating Rv2235/MT2294 interactions with other proteins in the mycobacterial proteome?

Investigating protein-protein interactions for Rv2235/MT2294 requires a multi-faceted approach combining both in vitro and in vivo methodologies. Given the limited specific information available about this protein, researchers should implement comprehensive interaction screening strategies.

Affinity purification coupled with mass spectrometry (AP-MS) represents a powerful initial approach. This methodology involves expressing Rv2235 with an affinity tag (e.g., FLAG, HA, or His tag), followed by pulldown of the protein along with its interaction partners from mycobacterial lysates. Subsequent mass spectrometry analysis can identify co-purified proteins. This approach has been successfully applied to other mycobacterial proteins, as demonstrated in case studies with proteins like NME1 and DNM2 .

Complementary to AP-MS, researchers should employ reciprocal co-immunoprecipitation experiments to validate key interactions, similar to the two-way co-immunoprecipitation approach used to confirm NME-DNM2 interaction . For comprehensive interaction mapping, yeast two-hybrid or bacterial two-hybrid screening can identify direct binary interactions.

More advanced techniques include proximity-dependent biotin identification (BioID) or proximity ligation assay (PLA), which can capture transient or weak interactions in their native cellular context. Researchers should also consider fluorescence resonance energy transfer (FRET) or bimolecular fluorescence complementation (BiFC) for visualizing interactions in live mycobacterial cells.

For validation of specific interactions, surface plasmon resonance (SPR) or isothermal titration calorimetry (ITC) can provide quantitative binding parameters including affinity constants and thermodynamic profiles. Finally, cross-linking mass spectrometry (XL-MS) can map specific interaction interfaces between Rv2235 and its binding partners at the amino acid level.

How can gene knockout or knockdown approaches be optimized for studying Rv2235/MT2294 function in M. tuberculosis?

Optimizing gene knockout or knockdown approaches for Rv2235/MT2294 requires careful methodology selection based on the specific research questions and the essential or non-essential nature of the gene. For M. tuberculosis genes, several complementary approaches have proven effective.

CRISPR-Cas9 systems adapted for mycobacteria offer precise genome editing capabilities for creating clean knockouts. Researchers should design guide RNAs targeting unique regions of Rv2235 with minimal off-target effects, followed by homology-directed repair to introduce specific modifications. For potentially essential genes like Rv2235, conditional knockout systems such as tetracycline-inducible promoters or degradation tags provide regulated expression control, allowing researchers to study the effects of protein depletion under controlled conditions.

CRISPRi (CRISPR interference) represents another valuable approach, using catalytically inactive Cas9 (dCas9) to block transcription without permanently altering the genome. This method can achieve significant knockdown (typically 50-95%) while avoiding potential issues with essential genes. Researchers should optimize guide RNA design to target the promoter region or early coding sequence of Rv2235 for maximal repression.

For stable knockdown, antisense RNA or RNA interference approaches can be employed, though their efficiency in mycobacteria may be variable. Regardless of the selected approach, comprehensive phenotypic characterization should include growth kinetics under various conditions, metabolic profiling, transcriptomic analysis, and specific assays related to respiratory function, given Rv2235's SURF1-like classification.

Complementation studies are essential to confirm that observed phenotypes result specifically from Rv2235 manipulation rather than polar effects or off-target impacts. This involves reintroducing the wild-type gene or variants (e.g., point mutants) to determine if phenotypes can be rescued.

What computational tools and databases are most useful for predicting Rv2235/MT2294 function based on sequence homology?

Functional prediction for uncharacterized proteins like Rv2235/MT2294 requires integration of multiple computational tools and databases focused on different aspects of protein function. Researchers should implement a systematic approach combining sequence-based, structure-based, and context-based prediction methods.

For sequence homology analysis, researchers should utilize BLAST (Basic Local Alignment Search Tool) against comprehensive databases like UniProt, RefSeq, and specialized mycobacterial databases like MycoBrowser. More sensitive homology detection tools like PSI-BLAST, HHpred, or HMMER can identify remote homologs that basic BLAST might miss, which is particularly valuable for proteins with low sequence conservation like Rv2235.

Domain architecture analysis using InterPro, Pfam, or CDD (Conserved Domain Database) can identify functional domains characteristic of SURF1 proteins or reveal unexpected domain combinations. Researchers should also analyze transmembrane topology using predictors like TMHMM or Phobius, given that SURF1 proteins typically contain membrane-spanning regions.

For evolutionary context, researchers should employ phylogenetic analysis to examine Rv2235's relationship with characterized SURF1 proteins across species. Tools like MEGA, PhyML, or MrBayes can construct robust phylogenetic trees to infer evolutionary relationships and potential functional conservation. Synteny analysis examining the conservation of gene order around Rv2235 across mycobacterial species can provide additional functional insights.

Structure-based function prediction, using tools like I-TASSER, Phyre2, or AlphaFold2, can generate three-dimensional models based on structural homologs, potentially revealing binding pockets or catalytic sites. These predictions should be complemented with functional site prediction using tools like ConSurf or 3DLigandSite to identify conserved residues potentially involved in protein function.

What is the role of Rv2235/MT2294 in mycobacterial energy metabolism and respiratory chain function?

Investigating the role of Rv2235/MT2294 in mycobacterial energy metabolism requires integrated biochemical, genetic, and physiological approaches. As a SURF1-like protein, Rv2235 may function in aspects of respiratory chain assembly or regulation, similar to the role of SURF1 in cytochrome c oxidase assembly in other organisms .

Researchers should begin with comparative metabolomic profiling of wild-type versus Rv2235 knockout or knockdown strains using techniques like liquid chromatography-mass spectrometry (LC-MS) to identify altered metabolites in central carbon metabolism and respiratory pathways. This should be complemented with measurement of key bioenergetic parameters including ATP/ADP ratios, NADH/NAD+ ratios, and membrane potential using fluorescent probes like TMRM (tetramethylrhodamine methyl ester).

Oxygen consumption rates measured using respirometry techniques such as Seahorse XF analyzers or Clark-type oxygen electrodes can provide direct evidence of respiratory chain function. These measurements should be conducted under various growth conditions (aerobic, microaerobic, nutrient limitation) and in the presence of specific respiratory inhibitors to pinpoint effects on particular branches of the respiratory chain.

For more detailed analysis, researchers should examine the assembly and activity of specific respiratory complexes. Activity assays for cytochrome c oxidase and other respiratory enzymes, combined with blue native PAGE to analyze intact respiratory complexes, can reveal defects in complex assembly or stability. Proteomic approaches can determine if the absence of Rv2235 alters the stoichiometry of respiratory complex components.

Gene expression analysis using RNA-seq or qRT-PCR should examine how Rv2235 deletion affects expression of other genes involved in energy metabolism and respiratory function, potentially revealing compensatory responses or regulatory connections. This approach has been successfully applied to study other mycobacterial genes involved in metabolic functions .

How does Rv2235/MT2294 expression change during different stages of M. tuberculosis infection?

Understanding the expression dynamics of Rv2235/MT2294 during infection requires methodologies that can capture gene expression across diverse infection stages and microenvironments. Researchers should implement both in vitro and in vivo approaches to comprehensively characterize expression patterns.

For in vitro analysis, researchers should employ quantitative RT-PCR to measure Rv2235 expression under conditions mimicking different aspects of infection, including hypoxia, nutrient limitation, acidic pH, nitrosative stress, and exposure to host immune factors. These controlled experiments provide baseline data on how specific environmental cues regulate Rv2235 expression. Gene expression should be normalized against stable reference genes such as polA or 16S rRNA, similar to approaches used for other mycobacterial genes .

More sophisticated in vitro models including infected macrophages, granuloma-like structures, or lung tissue explants provide more physiologically relevant contexts. RNA extraction from bacteria in these systems, though technically challenging, offers valuable insights into host-induced expression changes. Single-cell RNA-seq approaches can resolve heterogeneity in bacterial populations that bulk methods might miss.

For in vivo expression analysis, researchers should utilize animal models of tuberculosis infection (typically mouse or guinea pig) with sampling at different infection stages (early, acute, chronic, reactivation). Bacterial RNA can be isolated directly from infected tissues or from bacteria recovered through differential centrifugation. Due to the low abundance of bacterial RNA in host tissues, techniques like microbial enrichment RNA-seq or amplification-based approaches may be necessary.

Reporter strains carrying fluorescent or luminescent proteins under control of the Rv2235 promoter can provide real-time visualization of expression in vivo through intravital microscopy. For clinical relevance, researchers should analyze Rv2235 expression in bacteria isolated from human clinical samples, though this approach faces significant technical challenges.

Could targeting Rv2235/MT2294 represent a viable approach for developing new anti-tuberculosis drugs?

Evaluating Rv2235/MT2294 as a potential drug target requires a systematic approach to assess its suitability based on established target validation criteria. Researchers should investigate essentiality, druggability, and potential implications for host-pathogen interactions.

Target validation should begin with essentiality assessment using conditional knockout systems or CRISPRi to determine if Rv2235 is required for mycobacterial survival or virulence in vitro and in vivo. Depletion phenotypes should be characterized across different growth conditions relevant to infection, as essentiality may be condition-dependent. If complete knockout is not viable, researchers should determine the minimal expression levels required for survival, as partial inhibition by drugs may still be therapeutically valuable.

Druggability assessment requires structural characterization to identify potential binding pockets that could accommodate small molecules. Computational methods like SiteMap or DoGSiteScorer can predict druggable sites based on structural models or experimentally determined structures of Rv2235. High-throughput screening approaches using fragment-based or virtual screening methods can identify chemical starting points for inhibitor development.

For compounds showing activity against Rv2235, researchers should establish structure-activity relationships through medicinal chemistry optimization, guided by biochemical assays measuring compound binding and inhibition of protein function. Lead compounds must then be evaluated for mycobactericidal activity against both replicating and non-replicating M. tuberculosis, with minimum inhibitory concentration (MIC) determination using standardized methods like REMA .

Pharmacological validation requires demonstration that compounds targeting Rv2235 can achieve efficacy in animal models of tuberculosis infection. This includes evaluation of pharmacokinetic and pharmacodynamic properties, toxicity assessment, and confirmation that observed antimicrobial effects correlate with Rv2235 inhibition rather than off-target effects.

Throughout the drug development process, researchers should implement resistance studies to understand potential resistance mechanisms and frequency, as this information is critical for designing optimal therapeutic strategies and combination regimens.

What is the prevalence of Rv2235/MT2294 mutations in clinical M. tuberculosis isolates, and do they correlate with disease outcomes?

Investigating the prevalence and clinical significance of Rv2235/MT2294 mutations requires integration of genomic, clinical, and functional approaches. Researchers should implement comprehensive methodologies to identify mutations and correlate them with disease phenotypes.

To establish clinical correlations, researchers must collect comprehensive metadata for isolates, including patient demographics, treatment histories, immune status, and detailed clinical outcomes. Statistical analyses should examine associations between specific Rv2235 mutations and variables such as disease severity, treatment response, relapse rates, and mortality. Longitudinal studies tracking mutation emergence during treatment can provide insights into adaptive roles of Rv2235 mutations.

Functional characterization of identified mutations is essential to understand their biological impact. This involves introducing mutations into laboratory M. tuberculosis strains using techniques like recombineering or CRISPR-Cas9 editing, followed by phenotypic characterization including growth kinetics, metabolic profiling, and virulence assessment in cellular and animal models. Biochemical analysis of recombinant mutant proteins can determine how mutations affect protein stability, interactions, or function.

For mutations affecting gene expression rather than protein structure, researchers should employ reporter assays with wild-type and mutant promoters to quantify expression differences. RNA-seq analysis of clinical isolates carrying different Rv2235 variants can reveal broader transcriptional consequences of these mutations.

What are the main challenges in expressing and purifying functional Rv2235/MT2294 protein, and how can they be overcome?

Expression and purification of functional membrane proteins like Rv2235/MT2294 present significant technical challenges requiring specialized approaches. Researchers must address issues related to protein folding, stability, and functional state preservation.

One primary challenge is low expression levels or toxicity in conventional expression systems. To address this, researchers should explore specialized bacterial expression strains like C41(DE3) or C43(DE3) specifically designed for membrane protein expression. Codon optimization of the Rv2235 sequence for the expression host can improve translation efficiency. Additionally, fusion tags such as MBP (maltose-binding protein) or SUMO can enhance solubility and expression levels.

Controlling expression rate through reduced temperature (16-20°C) and lower inducer concentrations often improves proper folding by giving the protein more time to integrate into membranes. For particularly challenging cases, cell-free expression systems offer an alternative that avoids toxicity issues and allows direct incorporation into artificial membrane environments.

Membrane protein solubilization represents another major challenge. Researchers should conduct detergent screening using a panel of detergents with varying properties (e.g., DDM, LMNG, CHAPS) to identify conditions that maintain protein stability and function. Nanodiscs, amphipols, or styrene-maleic acid copolymer lipid particles (SMALPs) offer alternative solubilization approaches that better mimic the native membrane environment.

For purification, researchers should implement a multi-step strategy typically including initial immobilized metal affinity chromatography (IMAC) followed by size exclusion chromatography (SEC) to separate monomeric protein from aggregates. On-column detergent exchange during purification can help transition from harsh solubilization detergents to milder ones for functional studies.

Quality control is essential throughout the process. Dynamic light scattering and analytical ultracentrifugation can assess sample homogeneity and oligomeric state. Circular dichroism spectroscopy can confirm proper secondary structure formation. For functional validation, researchers should develop activity assays relevant to predicted SURF1-like functions, potentially measuring interactions with components of respiratory complexes.

What experimental design considerations are most important when investigating potential interactions between Rv2235/MT2294 and drugs or small molecules?

Investigating interactions between Rv2235/MT2294 and small molecules requires careful experimental design to ensure robust, reproducible results that accurately reflect biologically relevant interactions. Researchers should address aspects of assay development, validation, and data interpretation.

For initial screening, researchers should establish biochemical assays that directly measure Rv2235 activity or binding interactions. Given its classification as a SURF1-like protein potentially involved in respiratory complex assembly, assays might include measuring interaction with cytochrome c oxidase components or monitoring assembly of respiratory complexes. These assays should be optimized for high signal-to-noise ratio, reproducibility, and adaptability to higher-throughput formats.

When direct functional assays are unavailable for uncharacterized proteins like Rv2235, biophysical binding assays represent a valuable alternative. Surface plasmon resonance (SPR), microscale thermophoresis (MST), or differential scanning fluorimetry (DSF) can detect compound binding without requiring knowledge of protein function. Researchers should include positive and negative controls to validate assay performance and establish significance thresholds.

Compound libraries should be designed or selected based on research objectives. For target-based drug discovery, focused libraries based on known inhibitors of homologous proteins may prove more productive than diverse chemical libraries. Researchers should implement quality control measures for compound libraries, including solubility assessment, stability testing, and exclusion of pan-assay interference compounds (PAINS) that can generate false positives.

For promising hits, researchers must establish concentration-response relationships through titration experiments to determine parameters like IC50, Kd, or EC50. Orthogonal assays employing different detection technologies should confirm initial hits and eliminate technology-specific artifacts. Selectivity profiling against related proteins can identify compounds with desirable specificity profiles.

The translation to whole-cell assays is critical for antimicrobial applications. Researchers should correlate biochemical activity against Rv2235 with whole-cell antimycobacterial activity. Target engagement in cells can be assessed using methodologies like cellular thermal shift assays (CETSA) or photoaffinity labeling. For genetic validation, testing compounds against Rv2235 overexpression or knockdown strains can confirm on-target activity.

What are the key considerations for designing and interpreting experiments involving Rv2235/MT2294 knockout or overexpression in model systems?

For knockout studies, researchers must first confirm complete elimination of Rv2235 expression through multiple methods including PCR verification of genomic modification, RT-qPCR to confirm absence of transcript, and Western blotting to verify protein absence. If complete knockout is lethal, conditional systems using tetracycline-responsive promoters or degradation tags offer controlled depletion. Researchers should be aware that essentiality can be condition-dependent, necessitating testing across various growth conditions relevant to M. tuberculosis lifecycle.

Complementation experiments are critical for confirming phenotype specificity. These should include both wild-type Rv2235 reintroduction and selected mutants (e.g., point mutations in conserved residues) to identify critical functional regions. Complementation constructs should allow physiological expression levels, as overexpression can mask subtle phenotypes or create artificial effects.

For overexpression studies, researchers should implement titratable expression systems rather than constitutive high-level expression, which can cause non-specific effects through protein aggregation or disruption of stoichiometric complexes. Expression levels should be quantified relative to endogenous Rv2235 levels to interpret results in a physiologically relevant context.

Phenotypic characterization requires comprehensive approaches spanning multiple levels of biological organization. Growth phenotypes should be assessed across diverse conditions including different carbon sources, oxygen levels, and stress conditions. More specific assays related to predicted SURF1-like functions should measure parameters like respiratory activity, cytochrome c oxidase assembly/activity, and energy metabolism indicators (ATP levels, membrane potential).

Researchers should implement global approaches like transcriptomics, proteomics, or metabolomics to capture broader consequences of Rv2235 manipulation. This systems-level view can reveal compensatory mechanisms or unexpected pathway connections. For in vivo studies, researchers should assess multiple parameters including bacterial burden, histopathology, and survival in animal models, while considering potential differences in host-specific environments.

How might single-cell approaches be applied to understand the role of Rv2235/MT2294 in M. tuberculosis heterogeneity during infection?

Single-cell approaches offer powerful tools for investigating how Rv2235/MT2294 contributes to phenotypic heterogeneity in M. tuberculosis populations during infection. Researchers should implement complementary methodologies to capture gene expression, protein levels, and functional parameters at single-cell resolution.

For gene expression analysis, researchers can employ single-cell RNA sequencing (scRNA-seq) adapted for bacterial cells, though this presents technical challenges due to low RNA content. Newer approaches like Split-seq or Seq-Well offer higher throughput and sensitivity. To specifically track Rv2235 expression, researchers should develop fluorescent reporter constructs where a fluorescent protein like mCherry or GFP is expressed under control of the Rv2235 promoter. When combined with microfluidic systems or flow cytometry, these reporters allow dynamic tracking of expression in living cells.

Single-cell protein detection can be achieved using techniques like mass cytometry (CyTOF) with metal-conjugated antibodies against Rv2235 or epitope tags. For spatial context within infected tissues, multiplexed ion beam imaging (MIBI) or imaging mass cytometry can simultaneously detect multiple bacterial and host proteins while preserving tissue architecture. These approaches can reveal whether Rv2235 expression correlates with specific microenvironments within granulomas.

Functional heterogeneity can be investigated using activity-based reporters responsive to parameters like metabolic state, membrane potential, or respiratory activity. These reporters, when combined with Rv2235 expression markers, can establish correlations between Rv2235 levels and functional states at single-cell resolution. Microfluidic devices allowing controlled environmental perturbations can reveal how individual cells with different Rv2235 expression levels respond to stresses like antibiotics or immune factors.

For tracking bacterial subpopulations over time, genetic barcoding combined with Rv2235 reporter systems can link initial expression states to long-term fate during infection. This approach is particularly valuable for understanding whether Rv2235 expression correlates with persistence or reactivation phenotypes.

Data analysis for single-cell studies requires sophisticated computational approaches. Researchers should employ dimensionality reduction techniques like t-SNE or UMAP to visualize phenotypic clusters, trajectory inference methods to map developmental pathways, and correlation analyses to identify genes co-regulated with Rv2235 at single-cell resolution.

What role might Rv2235/MT2294 play in M. tuberculosis adaptation to host environments and stress conditions?

Understanding Rv2235/MT2294's role in adaptation to host environments requires integrating stress response studies with host-pathogen interaction models. Researchers should implement systematic approaches to characterize how Rv2235 contributes to bacterial survival under diverse stress conditions encountered during infection.

Researchers should first establish an expression profile of Rv2235 under stresses relevant to the host environment, including hypoxia, nutrient limitation, oxidative/nitrosative stress, acidic pH, and exposure to host antimicrobial peptides. Quantitative RT-PCR or RNA-seq analysis comparing wild-type and regulatory mutants can identify signaling pathways controlling Rv2235 expression during stress adaptation.

For functional characterization, researchers should compare survival of wild-type, Rv2235 knockout, and complemented strains under these stress conditions. Growth curve analysis, colony-forming unit (CFU) determination, and live/dead staining can quantify differential survival. Long-term stress adaptation studies involving extended exposure to sub-lethal stressors can reveal roles in persistent phenotypes rather than just acute stress responses.

Researchers should investigate stress-induced phenotypic changes at the biochemical level. Metabolomic profiling using techniques like liquid chromatography-mass spectrometry can identify metabolic reconfigurations associated with Rv2235 during stress adaptation. Proteomic approaches focusing on membrane protein complexes can determine if Rv2235 mediates stress-induced remodeling of respiratory machinery.

Macrophage infection models offer controlled systems to study host-relevant stresses. Researchers should compare intracellular survival of wild-type and Rv2235-modified strains in resting macrophages, activated macrophages, and under conditions of altered phagosomal maturation. Time-course transcriptomic analysis of intracellular bacteria can identify Rv2235-dependent adaptive responses to the changing phagosomal environment.

For more complex host environments, researchers should utilize advanced in vitro models like 3D granuloma models or lung tissue explants that better recapitulate in vivo conditions. These systems allow evaluation of how Rv2235 contributes to adaptation in heterogeneous microenvironments more representative of human disease than simple culture conditions or single-cell type infections.

How can systems biology approaches enhance our understanding of Rv2235/MT2294 function within the context of M. tuberculosis metabolic networks?

Systems biology approaches provide powerful frameworks for contextualizing Rv2235/MT2294 function within the broader metabolic and regulatory networks of M. tuberculosis. Researchers should implement multi-omics integration and computational modeling to develop comprehensive functional insights.

Multi-omics data generation should begin with comparative analysis of wild-type and Rv2235 mutant strains across multiple biological levels. Transcriptomics using RNA-seq can identify genes with altered expression in response to Rv2235 mutation, potentially revealing regulatory networks. Proteomics using mass spectrometry can detect changes in protein abundance and post-translational modifications, with particular focus on membrane proteome and respiratory complexes given Rv2235's potential role in these systems. Metabolomics can identify altered metabolic pathways, particularly those related to energy metabolism and respiration.

For network analysis, researchers should apply correlation networks and mutual information approaches to identify genes and proteins whose expression patterns correlate with Rv2235 across multiple conditions. Protein-protein interaction networks constructed from experimental data (Y2H, AP-MS) and computational predictions can position Rv2235 within its functional context. Differential network analysis comparing network structures between normal and stress conditions can reveal condition-specific roles of Rv2235.

Genome-scale metabolic models (GEMs) of M. tuberculosis can be leveraged to predict the systemic impact of Rv2235 perturbation. Researchers should perform flux balance analysis (FBA) simulations with constraints derived from experimental data to predict metabolic consequences of Rv2235 deletion or overexpression. These predictions should be experimentally validated through techniques like 13C metabolic flux analysis to measure actual flux distributions.

Integration of multi-omics data with regulatory information can be achieved through approaches like gene regulatory network reconstruction, which can identify transcription factors controlling Rv2235 expression and genes co-regulated with it. More advanced modeling approaches like ordinary differential equation (ODE) models can capture the dynamics of Rv2235-associated processes, while Boolean network models can represent regulatory logic governing Rv2235 function in different conditions.

For translational relevance, researchers should implement drug target identification algorithms using network approaches such as vulnerability analysis or synthetic lethality prediction to evaluate Rv2235's potential as a drug target in the context of system-level understanding. Comparative systems analysis across multiple mycobacterial species can provide evolutionary context for Rv2235 function and its conservation across pathogenic and non-pathogenic mycobacteria.

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.