KEGG: aga:AgaP_AGAP008883
STRING: 7165.AGAP008883-PA
Anamorsin homolog (AGAP008883) in Anopheles gambiae functions as a component of the cytosolic iron-sulfur (Fe-S) protein assembly (CIA) machinery. The protein is required for the maturation of extramitochondrial Fe-S proteins and acts as part of an electron transfer chain during an early step of cytosolic Fe-S biogenesis, facilitating the de novo assembly of [4Fe-4S] clusters on the cytosolic Fe-S scaffold complex . Together with a diflavin oxidoreductase, it contributes to the assembly of the diferric tyrosyl radical cofactor of ribonucleotide reductase (RNR), likely by providing electrons for reduction during radical cofactor maturation in the catalytic small subunit.
Similar to its homologs in other organisms, AGAP008883 likely plays a critical role in cellular iron homeostasis and oxidative stress responses, which may be particularly relevant to understanding the vector biology of A. gambiae in relation to Plasmodium infection and transmission.
Based on research experiences with recombinant A. gambiae proteins, multiple expression systems have been successfully employed for AGAP008883, each with specific advantages:
| Expression System | Advantages | Limitations | Yield | Applications |
|---|---|---|---|---|
| E. coli | High yield, cost-effective, rapid production | Limited post-translational modifications, potential for inclusion bodies | 10-20 mg/L | Structural studies, antibody production |
| Yeast (K. lactis) | Better protein folding, some post-translational modifications | Lower yield than E. coli, longer production time | 2-5 mg/L | Functional assays, protein-protein interaction studies |
| Baculovirus | Insect-specific post-translational modifications, proper folding | Higher cost, technical complexity | 1-10 mg/L | Enzymatic assays, structural biology |
| Mammalian cell | Most complete post-translational modifications | Highest cost, lowest yield, complex protocols | 0.5-2 mg/L | In vitro activity assays requiring native conformation |
For most applications, bacterial expression in E. coli remains the method of choice, particularly when followed by solubilization from inclusion bodies using established protocols . For functional studies where proper folding and post-translational modifications are critical, insect cell expression systems may be more appropriate.
The protein should be expressed with appropriate purification tags (His, GST, or MBP) and purified using affinity chromatography followed by size exclusion chromatography to obtain high purity preparations suitable for downstream applications .
When designing experiments to study AGAP008883 function in vivo, several methodological approaches should be considered:
RNA Interference (RNAi) Approach:
Design sequence-specific dsRNA targeting AGAP008883
Assess phenotypic effects on iron homeostasis, oxidative stress response, and potential impact on vector competence
Include appropriate controls (non-targeting dsRNA) and time-course analysis (knockdown typically peaks 2-7 days post-injection)
CRISPR-Cas9 Gene Editing:
Design guide RNAs specific to AGAP008883 with minimal off-target effects
Implement the GAL4/UAS system for conditional expression/knockdown
Include molecular verification of editing through sequencing
Monitor fitness parameters in modified mosquitoes
Transgenic Overexpression:
Design constructs with tissue-specific promoters (midgut, salivary gland, fat body)
Implement xenotransgenic approaches for cross-species validation
Verify expression levels through qPCR and protein quantification
Experimental Controls:
Include proper controls for mosquito age, rearing conditions, and genetic background
Design experiments with appropriate replication (minimum n=30 per condition)
Implement blinding when assessing phenotypes
Consider genetic background effects when using different A. gambiae strains
A comprehensive experimental design should incorporate multiple complementary approaches to fully elucidate AGAP008883 function in the context of mosquito biology and vector competence.
Investigating AGAP008883's role in iron metabolism requires a multifaceted experimental approach:
1. Dietary Iron Manipulation Studies:
Design feeding regimens with controlled iron concentrations (deficient, normal, excess)
Monitor AGAP008883 expression levels in response to dietary changes using qRT-PCR
Measure iron content in tissues using ferrozine assay or ICP-MS
Assess expression of iron regulatory genes (ferritin, transferrin) as controls
2. Blood Meal Response Analysis:
Compare AGAP008883 expression before and after blood feeding at multiple time points (3h, 24h, 48h post-feeding)
Correlate with heme metabolism genes and oxidative stress markers
Include sugar-fed controls to distinguish blood-specific effects
3. Knockdown Impact Assessment:
Implement RNAi-mediated silencing of AGAP008883
Measure iron distribution using Prussian blue staining or ferrozine assay
Assess impact on Fe-S cluster containing enzymes (aconitase, xanthine oxidase)
Evaluate oxidative stress markers (lipid peroxidation, protein carbonylation)
4. Protein-Protein Interaction Studies:
Identify binding partners using co-immunoprecipitation followed by mass spectrometry
Validate interactions with known CIA components using pull-down assays
Map interaction domains through truncation mutants
Quantify binding affinities using surface plasmon resonance
For statistical analysis, implement ANOVA for multi-condition comparisons with post-hoc Tukey's test, and use appropriate non-parametric tests when data doesn't meet normality assumptions. Sample sizes should be calculated based on preliminary data to achieve statistical power of at least 0.8.
Optimizing experimental design for protein interaction studies with AGAP008883 requires careful consideration of multiple factors:
In Vitro Approaches:
Yeast Two-Hybrid Screening
Construct bait plasmids containing full-length and domain-specific fragments of AGAP008883
Screen against A. gambiae cDNA libraries
Include appropriate controls (empty vectors, known interactors)
Validate positive hits with secondary assays
Pull-Down Assays
Express recombinant AGAP008883 with different tags (His, GST, MBP)
Use mosquito tissue lysates as prey
Implement stringent washing conditions to minimize false positives
Confirm results with reverse pull-downs using identified partners
Surface Plasmon Resonance
Immobilize purified AGAP008883 on sensor chips
Test binding kinetics with purified candidate partners
Determine association/dissociation constants
Evaluate effects of oxidation state and Fe-S cluster presence
In Vivo Approaches:
Co-Immunoprecipitation
Generate specific antibodies against AGAP008883
Prepare tissue lysates under native conditions
Include appropriate negative controls (pre-immune serum, non-relevant antibodies)
Confirm reciprocal co-IPs with partner antibodies
Proximity Ligation Assay
Design specific antibodies for AGAP008883 and candidate partners
Visualize interactions in situ in mosquito tissues
Quantify interaction signals across different tissues and conditions
Experimental Design Considerations:
Use factorial design to test multiple variables (protein concentration, buffer conditions, temperature)
Include biological replicates (minimum n=3) for each condition
Implement positive and negative controls for each experiment
Consider the natural oxidation state of the protein when designing buffer systems
Following these methodological approaches will maximize the likelihood of detecting authentic protein interactions while minimizing false positives and artifacts.
Distinguishing between direct and indirect effects of AGAP008883 in oxidative stress response requires sophisticated experimental design and careful controls:
1. Temporal Analysis Design:
Implement time-course experiments following AGAP008883 knockdown or overexpression
Monitor oxidative stress markers at short intervals (1h, 3h, 6h, 12h, 24h)
Graph temporal relationships between AGAP008883 levels and downstream effects
Primary effects typically manifest earlier than secondary consequences
2. Dose-Response Relationship Analysis:
Create mosquito lines with varying levels of AGAP008883 expression (RNAi with different efficiencies or inducible expression systems)
Measure correlation between AGAP008883 levels and oxidative stress parameters
Direct effects typically show stronger dose-response relationships than indirect effects
Fit data to appropriate mathematical models (linear, sigmoidal, etc.)
3. Pathway Inhibition Strategy:
Selectively inhibit potential intermediate pathways
If AGAP008883 effects persist despite pathway inhibition, direct action is supported
If effects are blocked, the inhibited pathway likely mediates AGAP008883 action
Include appropriate pharmacological controls
4. Reconstitution Experiments:
Purify recombinant AGAP008883 protein
Test direct effects in cell-free systems with isolated cellular components
Design in vitro assays measuring specific activities (Fe-S transfer, ROS scavenging)
Compare with known direct-acting antioxidant proteins
5. Protein Domain Mutation Analysis:
Generate AGAP008883 variants with mutations in functional domains
Test which domains are required for oxidative stress protection
Correlate specific biochemical activities with stress response phenotypes
Design mutations that separate Fe-S assembly function from other potential roles
Implementing these approaches in combination provides robust evidence to distinguish direct from indirect effects, critical for accurately defining AGAP008883's role in mosquito oxidative stress response.
Investigating AGAP008883's potential role in vector competence requires integrated approaches spanning molecular, cellular, and organismal levels:
1. Infection Studies with Gene Manipulation:
Generate AGAP008883 knockdown and overexpression mosquitoes
Challenge with Plasmodium berghei (rodent model) and P. falciparum (human parasite)
Measure infection parameters:
Oocyst prevalence and intensity
Sporozoite loads in salivary glands
Transmission efficiency to naive hosts
Include appropriate controls (age-matched, same genetic background)
2. Tissue-Specific and Temporal Expression Analysis:
Target expression modification in midgut, hemocytes, and salivary glands
Implement blood-meal inducible promoters for temporal control
Measure tissue-specific effects on parasite development stages
3. Molecular Pathway Analysis:
Perform transcriptomic analysis comparing wild-type and AGAP008883-modified mosquitoes during infection
Identify differentially expressed immune genes
Measure oxidative stress parameters during infection progression
Assess iron distribution in tissues during infection
4. Biochemical Interaction Studies:
Test direct interaction between purified AGAP008883 and Plasmodium proteins
Investigate AGAP008883 localization during infection using immunofluorescence
Determine if Plasmodium infection alters AGAP008883 expression or activity
Measure parasite iron acquisition in the presence/absence of AGAP008883
5. Field-Relevant Approaches:
Test multiple A. gambiae strains with different genetic backgrounds
Include environmental variables (temperature, humidity) in experimental design
Consider microbiome interactions through controlled colonization experiments
Validate laboratory findings with field-derived mosquito populations
This comprehensive approach will provide robust evidence for AGAP008883's potential role in vector competence while accounting for biological variability and environmental factors relevant to disease transmission dynamics.
1. Genetic Background Control:
Use isogenic lines when performing genetic modifications
Backcross modified lines to parental strain for at least 5 generations
Include multiple independent transgenic/knockdown lines in analysis
Implement appropriate genetic controls (e.g., non-targeting RNAi, empty vector transgenics)
2. Off-Target Effect Mitigation:
Design multiple RNAi constructs targeting different regions of AGAP008883
Validate specificity through transcriptome analysis
Perform rescue experiments with RNAi-resistant transgenes
Use CRISPR-Cas9 with multiple guide RNAs to confirm phenotypes
3. Life History Trait Assessment:
Comprehensively measure:
Development time
Adult longevity
Reproductive output
Blood-feeding behavior
General fitness parameters
Determine if phenotypes are specific or result from general health impairment
4. Environmental Variable Control:
Standardize:
Rearing temperature (27±1°C)
Humidity (75±5%)
Photoperiod (12:12 light:dark)
Diet composition and feeding regimen
Experimental timing relative to mosquito age
Include environmental measurements as covariates in statistical models
5. Statistical Approaches for Confounding Control:
Implement factorial designs to test interaction effects
Use multivariate analysis to control for correlated variables
Apply propensity score matching when randomization is imperfect
Perform sensitivity analysis to test robustness of findings
6. Reporting and Transparency:
Document all experimental conditions in detail
Report negative and inconclusive results
Include comprehensive methods for replication
Share raw data in public repositories
By systematically addressing these potential confounding factors, researchers can substantially increase confidence that observed phenotypes are specifically attributable to AGAP008883 modification rather than experimental artifacts or secondary effects.
Post-translational modifications (PTMs) of AGAP008883 are likely critical for its function in Fe-S cluster assembly. The following methodological approaches are recommended for comprehensive PTM analysis:
1. Mass Spectrometry-Based Approaches:
Implement bottom-up proteomics with tryptic digestion
Use complementary fragmentation methods (CID, ETD, HCD) for comprehensive coverage
Apply titanium dioxide enrichment for phosphorylation analysis
Utilize HILIC fractionation for glycopeptide enrichment
Employ targeted MRM for quantitative analysis of specific modifications
Sample Preparation Considerations:
Extract protein under non-reducing conditions to preserve disulfide bonds
Use multiple proteases (trypsin, chymotrypsin, AspN) for improved sequence coverage
Include phosphatase inhibitors to preserve phosphorylation states
Minimize oxidation during sample handling with argon overlay
2. Site-Directed Mutagenesis Strategy:
Identify putative PTM sites through bioinformatic prediction and MS validation
Generate site-specific mutants (S→A for phosphorylation, C→S for disulfide bonds)
Compare functional activity of wild-type and mutant proteins
Create comprehensive mutation panels to assess combinatorial effects
3. Specific PTM Analysis Methods:
| PTM Type | Detection Method | Quantification Approach | Functional Validation |
|---|---|---|---|
| Phosphorylation | Phos-tag SDS-PAGE, ProQ Diamond staining | SILAC/TMT labeling, MRM | Phosphomimetic mutations (S→D/E) |
| Disulfide bonds | Non-reducing SDS-PAGE, Diagonal electrophoresis | MS with differential alkylation | C→S mutations, reduction sensitivity |
| Fe-S coordination | UV-vis spectroscopy, EPR | Iron quantification, Mössbauer spectroscopy | Ligand mutations, reconstitution assays |
| Glycosylation | Lectin blotting, PNGase F treatment | HILIC-MS | Tunicamycin treatment, N→Q mutations |
4. Expression System Selection for PTM Studies:
Use insect cell expression systems (Sf9, High Five) for most authentic PTM profiles
Compare modifications across expression systems to identify critical PTMs
Consider native purification from A. gambiae as reference standard
Implement in vitro modification with recombinant enzymes for mechanistic studies
These methodological approaches will provide comprehensive insights into the PTM landscape of AGAP008883 and their functional significance in mosquito biology.
Investigating AGAP008883 function across insecticide-resistant and susceptible Anopheles strains presents specific methodological challenges that require careful experimental design:
1. Genetic Background Variability:
Challenge: Resistant and susceptible strains often differ in multiple genetic loci beyond resistance genes
Solution: Create near-isogenic lines through backcrossing resistant strains to susceptible backgrounds
Methodology: Implement at least 5-10 generations of backcrossing with molecular marker selection
Validation: Confirm genetic similarity through whole-genome sequencing or SNP panel analysis
2. Resistance Mechanism Heterogeneity:
Challenge: Different resistance mechanisms (target-site, metabolic, cuticular) may interact differently with AGAP008883 function
Solution: Characterize specific resistance mechanisms in each strain using bioassays and molecular diagnostics
Methodology: Implement WHO tube assays, biochemical assays for enzyme activity, and molecular genotyping for kdr, rdl, and ace-1 mutations
Analysis: Stratify results by resistance mechanism type and intensity
3. Tissue-Specific Expression Differences:
Challenge: Resistance may alter tissue-specific expression patterns of multiple genes
Solution: Implement tissue-specific transcriptomic and proteomic profiling
Methodology: Use tissue microdissection followed by qRT-PCR or RNA-Seq for expression analysis
Controls: Include housekeeping genes with stable expression across resistant and susceptible strains
4. Physiological State Standardization:
Challenge: Resistant mosquitoes may differ in life history traits affecting experimental outcomes
Solution: Standardize age, feeding status, and physiological condition
Methodology: Use tightly synchronized cohorts, controlled feeding protocols, and standardized rearing conditions
Measurements: Monitor key physiological parameters (body size, weight, nutritional reserves)
5. Interaction with Resistance-Conferring Proteins:
Challenge: Resistance proteins (e.g., P450s, GSTs) may functionally interact with AGAP008883
Solution: Implement protein-protein interaction studies comparing resistant and susceptible variants
Methodology: Use co-immunoprecipitation, proximity ligation assays, and yeast two-hybrid screens
Validation: Confirm interactions through in vitro reconstitution with purified components
6. Data Analysis and Interpretation Frameworks:
Challenge: Complex interactions between resistance status and AGAP008883 function
Solution: Implement multifactorial experimental designs and appropriate statistical models
Methodology: Use mixed-effects models incorporating resistance status, genetic background, and environmental factors as variables
Validation: Perform independent validation in multiple strain comparisons
Addressing these methodological challenges systematically will enable robust comparisons of AGAP008883 function between resistant and susceptible strains, potentially revealing novel connections between iron metabolism and insecticide resistance mechanisms.
Designing experiments to investigate AGAP008883's interaction with the immune system during Plasmodium infection requires an integrated approach:
1. Temporal Immune Response Profiling:
Design time-course experiments sampling at key infection stages (3h, 24h, 3d, 7d, 14d post-infection)
Measure AGAP008883 expression alongside immune genes (TEP1, LRIM1, APL1C)
Implement both qRT-PCR for targeted analysis and RNA-Seq for global profiling
Compare patterns between susceptible and refractory mosquito strains
2. Immune Pathway Manipulation:
Perform dual knockdown experiments (AGAP008883 + key immune factors)
Assess epistatic relationships through phenotypic analysis
Focus on complement-like pathways involving TEP1, which is known to function with LRIM1/APL1C
Implement pathway-specific genetic manipulations using the GAL4/UAS system
3. Hemolymph Proteome Analysis:
Collect hemolymph at defined time points post-infection
Compare proteome profiles between wild-type and AGAP008883-modified mosquitoes
Identify changes in circulating immune factors
Quantify levels of iron-binding proteins and oxidative stress markers
4. Cellular Immune Response Assessment:
Analyze hemocyte numbers and types using flow cytometry
Measure phagocytic activity toward Plasmodium and model particles
Assess hemocyte-specific gene expression with single-cell RNA-Seq
Determine if AGAP008883 affects hemocyte differentiation or function
5. Oxidative Burst Measurement:
Design assays to quantify ROS production during infection
Compare oxidative response between control and AGAP008883-modified mosquitoes
Implement fluorescent probes for in vivo ROS visualization
Correlate oxidative burst intensity with parasite survival
6. Iron Homeostasis and Immune Function:
Manipulate dietary iron levels in combination with AGAP008883 modification
Measure impact on complement activation and melanization responses
Assess iron sequestration as an nutritional immunity mechanism
Determine if AGAP008883 mediates iron-dependent immune effector functions
Methodological Controls and Considerations:
Include appropriate genetic background controls
Implement parallel experiments with multiple Plasmodium species (P. berghei, P. falciparum)
Control for infection intensity through standardized feeding protocols
Consider environmental variables (temperature, microbiota) known to affect immune function