Recombinant Anopheles gambiae SAGA-associated factor 11 homolog (Sgf11)

Shipped with Ice Packs
In Stock

Description

Sgf11 in Yeast: Established Function and Mechanism

Sgf11 is a core component of the SAGA complex in Saccharomyces cerevisiae, where it functions within the deubiquitination module (DUBm) alongside Ubp8 (a ubiquitin hydrolase) and Sus1. Key findings from yeast studies include:

  • Role in H2B Deubiquitination: Sgf11 facilitates the removal of ubiquitin from histone H2B at lysine 123, enabling trans-tail methylation of H3 and promoting gene activation .

  • Structural Anchoring: Sgf11 interacts with Sgf73 (ATXN7 homolog) to anchor the DUBm to the SAGA complex. Loss of Sgf73 disrupts DUBm recruitment and impairs transcription elongation .

  • Conserved Zinc-Finger Domain: The Zn-Sgf11 domain is critical for structural integrity and functional interactions, though sequence conservation between species is limited .

FunctionYeast Sgf11 InteractionImpact of Mutation
H2B deubiquitinationUbp8, Sus1, Sgf73Reduced H3 methylation, gene activation defects
SAGA complex assemblySgf73, Sus1Loss of DUBm from SAGA, elongation defects
mRNA export couplingSus1, TREX2Cytoplasmic mislocalization of Sus1, export defects

Sgf11 in Anopheles gambiae: No Direct Evidence

The provided search results on Anopheles gambiae focus on immune response, insecticide resistance, and vector biology, with no mention of Sgf11 or the SAGA complex. Key findings include:

  • Immune Pathways: LRIM1/APL1C complexes stabilize TEP1, a C3-like protein critical for anti-Plasmodium defense .

  • Insecticide Resistance: Metabolic resistance involves CYP6P3 overexpression and kdr mutations (Vgsc alleles) .

  • Genomic Diversity: Population studies highlight introgression of kdr alleles between An. gambiae and An. coluzzii .

No data exists on homologs of Sgf11, Ubp8, or Sgf73 in Anopheles gambiae in the provided sources.

Potential Research Directions

Given the absence of direct evidence, future studies could explore:

  1. Ortholog Identification: Use bioinformatics tools to identify Sgf11 homologs in Anopheles genomes (e.g., Ag3.8 database ).

  2. Functional Conservation: Investigate whether Anopheles Sgf11 (if present) retains roles in chromatin remodeling or transcription elongation.

  3. Synthetic Biology Applications: Design recombinant Sgf11 for genetic manipulation, akin to CRISPR-based Y-chromosome shredding strategies .

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Consult your local distributor for precise delivery estimates.
Note: Standard shipping includes 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 collect the contents. Reconstitute the protein in sterile deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50%, which can be used as a guideline.
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 formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Upon receipt, store at -20°C/-80°C. Aliquot for multiple uses to prevent repeated freeze-thaw cycles.
Tag Info
The tag type is determined during the manufacturing process.
The tag type will be determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Sgf11; AGAP001393; SAGA-associated factor 11 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-182
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Anopheles gambiae (African malaria mosquito)
Target Names
Sgf11
Target Protein Sequence
MSENDGIHIE YVDEQELKRE FRRFMSDPET REKAANYLYE SLVDETILGI AYEVHHANKT GSGAAVEGEP EDSKPYTIVD QPDTDVFGSS NTKKAIDCHC PNCNRIVAAS RFAPHLEKCM GMGRNSSRIA SRRIANTRDV GTGNYFGGDE DDEDDADWSG EKRKKKISQV RTNGSKKNGK TS
Uniprot No.

Target Background

Function
Recombinant Anopheles gambiae SAGA-associated factor 11 homolog (Sgf11) is a component of the SAGA (Spt-Ada-Gcn5-acetyltransferase) complex, a multiprotein transcriptional regulatory complex. SAGA functions in chromatin remodeling, histone acetylation, and deubiquitination, ultimately activating transcription. Sgf11 participates in a SAGA subcomplex responsible for histone H2B deubiquitination. The SAGA complex, recruited to gene promoters by transcriptional activators, is essential for gene expression.
Database Links
Protein Families
SGF11 family
Subcellular Location
Nucleus.

Q&A

What is Anopheles gambiae Sgf11 and what is its role in the SAGA complex?

Sgf11 (SAGA-associated factor 11) is a protein component of the deubiquitination module (DUBm) within the SAGA chromatin-modifying complex in Anopheles gambiae. The SAGA complex possesses dual enzymatic activities: histone acetyltransferase activity and deubiquitination activity . The DUBm typically consists of Sgf11, ENY2, and Ubp8 (Nonstop) proteins, as observed in Drosophila . Sgf11 plays a crucial role in anchoring the DUBm to the SAGA complex, facilitating proper assembly and function. Within the context of SAGA, Sgf11 contributes to transcriptional regulation by modulating histone modifications, particularly H2B deubiquitination, which affects chromatin structure and accessibility for transcription machinery.

How is the SAGA complex assembled and what role does Sgf11 play in this process?

The SAGA complex assembly follows an ordered, directional pathway. Research in yeast has shown that core scaffolding components assemble first, followed by functional modules. The DUB module, containing Sgf11, appears to be integrated in the later stages of complex assembly. Evidence suggests that the incorporation of Tra1 (a SAGA component) precedes and may facilitate the integration of the DUB module containing Sgf11 .

The assembly process involves:

  • Core scaffold formation with Spt7 and other structural components

  • Integration of Spt20

  • Incorporation of Tra1

  • Assembly of the DUB module (including Sgf11)

Biochemical studies have demonstrated that Spt20 is required for both Tra1 and DUB module incorporation, while Tra1 stabilizes the DUB module but not Spt20 . This suggests a hierarchical assembly where Sgf11 integration depends on prior proper assembly of other components.

What expression systems are most effective for producing recombinant Anopheles gambiae Sgf11?

For recombinant expression of Anopheles gambiae Sgf11, several expression systems can be utilized, each with specific advantages:

Expression SystemAdvantagesLimitationsYield Potential
E. coliFast growth, simple media, cost-effectiveMay lack proper folding for eukaryotic proteinsMedium-High
Insect cells (Sf9, High Five)Native-like post-translational modificationsLonger production time, higher costHigh
Yeast (S. cerevisiae, P. pastoris)Eukaryotic processing, scalableLengthy optimization requiredMedium
Cell-free systemsRapid production, avoids toxicity issuesExpensive, lower yieldLow-Medium

For functional studies, insect cell expression systems often provide the best balance between proper protein folding and yield for mosquito proteins. Codon optimization for the expression host is crucial for improving yield. When co-expression with other DUBm components is needed for proper complex formation, baculovirus-based multicistronic vectors in insect cells represent the preferred approach.

What purification strategies work best for recombinant Sgf11?

Purification of recombinant Sgf11 typically employs a multi-step approach:

  • Initial capture: Affinity chromatography using a fusion tag (6xHis, GST, or MBP) is the most common first step. His-tagged Sgf11 can be purified using immobilized metal affinity chromatography (IMAC) with Ni-NTA or Co2+ resins.

  • Intermediate purification: Ion exchange chromatography (typically anion exchange) helps remove contaminants based on charge differences.

  • Polishing: Size exclusion chromatography separates any aggregates or degradation products from the correctly folded monomeric protein.

For structural studies or when high purity is required, additional steps such as heparin affinity chromatography may be beneficial due to Sgf11's nucleic acid binding properties. When co-purifying Sgf11 as part of the DUB module, tandem affinity purification using tags on different subunits can improve complex homogeneity.

How can researchers verify the functionality of recombinant Sgf11?

To verify recombinant Sgf11 functionality:

  • Complex formation assay: Assess the ability of recombinant Sgf11 to form complexes with other DUBm components (Ubp8/Nonstop and ENY2) using pull-down assays, size-exclusion chromatography, or native PAGE.

  • Deubiquitination activity assay: Measure the deubiquitinase activity of reconstituted DUBm containing recombinant Sgf11 using fluorogenic substrates or ubiquitinated histone H2B as substrates.

  • Chromatin binding assay: Evaluate binding of recombinant Sgf11-containing complexes to nucleosomes or chromatin using electrophoretic mobility shift assays (EMSA) or pull-down experiments.

  • Complementation assays: Introduce recombinant Sgf11 into Sgf11-depleted mosquito cells and assess rescue of transcriptional phenotypes.

  • Protein-protein interaction studies: Use techniques such as yeast two-hybrid, biolayer interferometry, or isothermal titration calorimetry to verify interactions with known binding partners.

What are the most effective gene editing approaches for studying Sgf11 function in Anopheles gambiae?

Several cutting-edge gene editing approaches can be employed for Sgf11 functional studies in Anopheles gambiae:

  • CRISPR-Cas9 embryonic injection: This direct approach involves microinjection of CRISPR-Cas9 components into mosquito embryos to achieve knockout or precise modifications of the Sgf11 gene. While technically challenging, it provides the most straightforward method for generating stable transgenic lines .

  • ReMOT Control (Receptor-mediated Ovary Transduction of Cargo): This innovative approach delivers gene-editing components to developing oocytes via receptor-mediated endocytosis after adult female injection. Recent studies in Anopheles sinensis have demonstrated high editing efficiency using this method, with significant benefits for laboratories unable to perform embryo injections . For Sgf11 studies, this approach could allow more efficient generation of knockout or knockin lines.

  • RNAi-mediated gene silencing: For temporary knockdown of Sgf11, RNAi remains a valuable approach. dsRNA injection into adult mosquitoes followed by phenotypic analysis has been successfully used for functional studies of salivary gland genes in A. gambiae .

The choice between these methods depends on research objectives:

  • For permanent genetic modifications, CRISPR-Cas9 or ReMOT Control are preferable

  • For rapid, transient analysis, RNAi offers a more accessible approach

  • For precise modifications (e.g., introducing specific mutations or tags), CRISPR-Cas9 with homology-directed repair is optimal

How can researchers characterize the transcriptional effects of Sgf11 disruption in Anopheles gambiae?

Characterizing transcriptional changes following Sgf11 disruption requires a multi-faceted approach:

  • RNA-Seq analysis: Perform comprehensive transcriptome profiling comparing wild-type and Sgf11-depleted mosquitoes. This approach has been successfully used to identify differentially expressed genes in Anopheles gambiae under various conditions .

  • ChIP-Seq analysis: Map genome-wide changes in histone modifications, particularly H2B ubiquitination levels, to correlate with transcriptional changes. This provides mechanistic insights into how Sgf11 disruption affects chromatin structure.

  • ATAC-Seq or MNase-Seq: Evaluate changes in chromatin accessibility resulting from Sgf11 depletion to identify regions where SAGA-dependent chromatin remodeling is compromised.

  • PRO-Seq or GRO-Seq: Measure nascent transcription to distinguish direct transcriptional effects from secondary effects on mRNA stability.

  • Allele-specific expression analysis: Crossing mosquitoes from different genetic backgrounds allows quantification of cis- versus trans-regulatory effects, as demonstrated in recent A. gambiae studies . This approach can help determine whether Sgf11 effects are primarily through local chromatin changes or broader regulatory networks.

For data analysis, integrate multiple datasets to identify high-confidence Sgf11-dependent genes, focusing on:

  • GO term enrichment

  • Pathway analysis

  • Motif discovery in affected promoters

  • Correlation with known SAGA binding sites

What is the role of Sgf11 in mosquito immunity and pathogen transmission?

The role of Sgf11 in mosquito immunity and vector competence remains largely unexplored, but can be investigated through several approaches:

  • Challenge experiments: Sgf11-depleted mosquitoes should be challenged with Plasmodium parasites and other pathogens to assess changes in infection intensity and prevalence.

  • Immune gene expression analysis: Examination of immune gene expression in Sgf11-depleted mosquitoes before and after immune challenge can reveal regulatory roles. This is particularly important as studies in Anopheles stephensi have shown that disruption of immune genes like LRIM1 can significantly alter the midgut microbiome composition and affect vector competence for Plasmodium .

  • Microbiome analysis: Characterization of the microbiome in Sgf11-depleted mosquitoes is essential, as alterations in the microbiome can indirectly impact pathogen transmission. In A. stephensi, LRIM1 knockout led to increased bacterial load and altered microbiome composition, ultimately affecting Plasmodium development .

  • Functional pathway analysis: Investigation of how Sgf11 regulation intersects with known immune signaling pathways (Toll, IMD, JAK-STAT) can provide mechanistic insights.

The potential connection between Sgf11 and immunity is significant because:

  • Chromatin modifiers often regulate stress and immune responses

  • Transcriptional regulation of immune genes is critical for mosquito vector competence

  • SAGA complex components may interact with immune signaling pathways

How does Sgf11 contribute to insecticide resistance mechanisms in Anopheles gambiae?

Investigating Sgf11's potential role in insecticide resistance requires:

  • Comparative expression analysis: Compare Sgf11 expression levels between insecticide-resistant and susceptible mosquito strains. Recent research has revealed that metabolic resistance in A. gambiae often involves overexpression of detoxification enzymes such as P450s .

  • Regulatory network analysis: Determine whether Sgf11-containing complexes regulate the expression of known insecticide resistance genes. This could involve:

    • ChIP-seq of SAGA components at promoters of resistance genes

    • Analysis of histone modifications at these loci

    • Assessment of chromatin accessibility changes following Sgf11 depletion

  • Genetic association studies: Analyze whether polymorphisms in Sgf11 or its binding sites correlate with resistance phenotypes across mosquito populations.

  • Functional validation: Test whether Sgf11 depletion affects mosquito survival upon insecticide exposure. Research in A. gambiae has established methodologies for WHO tube assays that could be adapted for this purpose .

  • Allele-specific expression analysis: Examine whether resistance-associated genes show differential allelic expression that might be regulated by Sgf11-dependent mechanisms. Recent work in A. gambiae has implemented methods to study allele-specific expression differences .

The epigenetic regulation of insecticide resistance is an emerging field, and characterizing Sgf11's contribution could provide novel insights into resistance mechanisms and potential intervention strategies.

What protein-protein interactions does Sgf11 form within and outside the SAGA complex?

Understanding Sgf11's interactome is crucial for elucidating its functional roles:

  • Co-immunoprecipitation coupled with mass spectrometry: This approach can identify stable and transient Sgf11 interaction partners in mosquito cells. Studies in yeast have used similar approaches to map SAGA complex interactions .

  • Proximity labeling approaches: BioID or TurboID fusion proteins can identify proximal proteins in living cells, potentially revealing transient or context-specific interactions.

  • Yeast two-hybrid screening: This can be used to identify direct binary interactions between Sgf11 and other proteins.

  • Structural studies: Cryo-EM or X-ray crystallography of purified complexes can provide atomic-level details of Sgf11 interactions within the SAGA complex.

Based on research in other organisms, expected interaction partners include:

  • Core DUBm components (Ubp8/Nonstop and ENY2)

  • Sgf73, which anchors the DUBm to SAGA

  • Potentially other chromatin-associated factors

These interactions are likely functionally important as:

  • Studies in yeast have shown that the assembly of SAGA follows an ordered pathway where Spt20 anchors Tra1, which then stabilizes the DUB module

  • The proper assembly of these complexes is essential for their chromatin-modifying activities

  • Disrupting specific protein-protein interactions could affect specific SAGA functions while preserving others

What is the optimal RNAi protocol for Sgf11 knockdown in Anopheles gambiae?

For effective RNAi-mediated knockdown of Sgf11 in Anopheles gambiae:

  • dsRNA design:

    • Target unique regions of Sgf11 (avoiding conserved domains shared with other proteins)

    • Design 300-500 bp fragments for efficient processing

    • Include GFP dsRNA as a negative control

  • dsRNA synthesis protocol:

    • PCR-amplify target region with primers containing T7 promoter sequences

    • Perform in vitro transcription using T7 RNA polymerase

    • Anneal sense and antisense RNA strands by heating and slow cooling

    • Confirm quality by gel electrophoresis

  • Injection methodology:

    • Inject 69 nl of dsRNA (3 μg/μl) into the thorax of cold-anesthetized 3-day-old female mosquitoes

    • Allow 4 days for efficient knockdown before phenotypic analysis

    • Confirm knockdown efficiency by RT-qPCR

  • Phenotypic assessment:

    • For blood-feeding related studies, starve mosquitoes from sugar for 4-5 hours before conducting behavioral assays

    • Define probing time as the period from initial mouthpart insertion to blood engorgement

    • Track multiple metrics including probing time, feeding capacity, and post-feeding behaviors

For statistical robustness, use at least three independent biological replicates with 20-30 mosquitoes per group. The Mann-Whitney test is appropriate for analyzing probing time data as demonstrated in previous studies .

How can CRISPR-Cas9 be optimized for Sgf11 gene editing in Anopheles gambiae?

Optimization of CRISPR-Cas9 for Sgf11 editing requires:

  • Guide RNA design:

    • Select targets with minimal off-target potential using mosquito-specific prediction tools

    • Target early exons to ensure functional disruption

    • Design multiple gRNAs to increase editing efficiency

    • Preferentially target conserved functional domains

  • Delivery methods:

    • Embryo microinjection: Inject Cas9 protein (300-500 ng/μl) complexed with gRNAs (100-150 ng/μl) into embryos within 1 hour of oviposition

    • ReMOT Control: For labs without embryo injection capabilities, this method delivers gene-editing components to developing oocytes after adult female injection

  • Donor template design (for knock-in or precise editing):

    • Include ~800-1000 bp homology arms flanking the modification site

    • Incorporate silent mutations in the PAM site to prevent re-cutting after HDR

    • Consider including selectable markers or fluorescent reporters

  • Screening strategies:

    • T7 endonuclease assay or heteroduplex mobility assay for initial detection

    • Direct sequencing to confirm modifications

    • Droplet digital PCR for precise quantification of editing efficiency

Recent improvements in ReMOT Control have demonstrated high editing efficiency in Anopheles sinensis, with benefits for laboratories without embryo injection capabilities . These approaches can be adapted for Sgf11 editing in A. gambiae.

What are the best approaches for studying Sgf11's role in chromatin modification?

To investigate Sgf11's chromatin-modifying functions:

  • Chromatin Immunoprecipitation (ChIP):

    • Develop specific antibodies against A. gambiae Sgf11 or use epitope-tagged versions

    • Perform ChIP-seq to map genome-wide binding sites

    • Compare with maps of histone modifications, particularly H2B ubiquitination and H3 acetylation

    • Analyze binding site motifs to identify sequence preferences

  • Histone modification analysis:

    • Western blotting to assess global levels of H2B ubiquitination in Sgf11-depleted cells

    • ChIP-seq for H2Bub, H3ac, and other relevant modifications to map changes at specific loci

    • Mass spectrometry of histone modifications to identify novel targets

  • Nucleosome positioning and accessibility:

    • ATAC-seq to assess changes in chromatin accessibility

    • MNase-seq to map nucleosome positioning changes

    • DNase-seq to identify hypersensitive sites affected by Sgf11 depletion

  • Biochemical assays:

    • Reconstitute the DUB module with recombinant components including Sgf11

    • Perform in vitro deubiquitination assays using nucleosomal substrates

    • Test the impact of Sgf11 mutations on enzymatic activity

These approaches can reveal both genome-wide patterns and mechanistic details of how Sgf11 contributes to chromatin regulation in Anopheles gambiae.

What are the major challenges in studying Sgf11 function in Anopheles gambiae?

Researchers face several significant challenges when studying Sgf11 in Anopheles gambiae:

  • Technical limitations:

    • Limited availability of mosquito-specific antibodies for native protein detection

    • Challenges in maintaining laboratory colonies with genetic modifications

    • Lower efficiency of genetic manipulation tools compared to model organisms

    • Difficulty in performing embryonic injections requires specialized equipment and expertise

  • Biological complexity:

    • Potential redundancy or compensation within transcriptional regulatory networks

    • Life-stage and tissue-specific expression patterns requiring temporal control of genetic manipulations

    • Potential essential functions making complete knockout lethal

    • Differences in chromatin regulation between mosquitoes and model organisms where SAGA is better characterized

  • Resource constraints:

    • Fewer genetic tools compared to model organisms

    • Limited public databases specific to A. gambiae gene regulation

    • Higher costs associated with mosquito maintenance compared to other insects

  • Experimental design considerations:

    • Need for appropriate controls in crossing experiments to account for genetic background effects

    • Challenges in distinguishing direct from indirect effects in global transcriptional studies

    • Variability in microbiome composition potentially affecting experimental outcomes

Addressing these challenges requires innovative approaches that combine cutting-edge technology with mosquito-specific adaptations.

How might understanding Sgf11 function contribute to novel vector control strategies?

Investigating Sgf11 could contribute to vector control in several ways:

  • Gene drive applications:

    • If Sgf11 proves essential for mosquito reproduction or development, it could become a target for gene drive-based population suppression

    • Modification of Sgf11 regulatory networks might enable modulation of pathogen susceptibility

  • Identification of novel insecticide targets:

    • Understanding how Sgf11 contributes to insecticide resistance mechanisms could reveal new resistance biomarkers

    • Sgf11-dependent transcriptional networks might identify new targets for chemical control

  • Genetic control strategies:

    • Knowledge of Sgf11's role in reproductive processes could inform sterile insect technique approaches

    • Manipulation of Sgf11-dependent pathways might allow for sex-specific control mechanisms

  • Host-pathogen interaction insights:

    • If Sgf11 regulates immune gene expression, it could provide targets for boosting anti-Plasmodium immunity

    • Understanding how pathogens might manipulate host transcription via SAGA-dependent mechanisms could reveal intervention points

Recent functional genomics research in mosquito vectors has provided essential molecular targets for genetic control strategies . Detailed characterization of transcriptional regulators like Sgf11 expands our toolkit for developing next-generation vector control approaches that are species-specific and environmentally friendly.

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 2025 TheBiotek. All Rights Reserved.