Recombinant Anopheles gambiae Protein CLP1 homolog (cbc)

Shipped with Ice Packs
In Stock

Description

Introduction

The Anopheles gambiae Leucine-rich repeat (LRR) protein APL1 family consists of three independently transcribed genes: APL1A, APL1B, and APL1C . Among these, APL1C is pivotal for the mosquito's defense against the malaria parasite Plasmodium berghei . APL1C operates within the Rel1-Cactus immune signaling pathway, influencing both its transcript and protein levels .

Discovery and Function

Originally identified through genetic mapping, the APL1 region of the Anopheles gambiae chromosome showed a strong correlation with the outcome of malaria parasite infection . Further investigation revealed that silencing the APL1C gene negates the Rel1-mediated protection against P. berghei, underscoring its necessity in this defense mechanism . The APL1C mRNA levels are modulated by the Rel1/Cactus immune signaling pathway, where Rel1 positively regulates APL1C, and Cactus negatively regulates it .

Role in Mosquito Immunity

APL1C is essential for the mosquito's immune response against Plasmodium parasites . It functions as a required transducer of Rel1-dependent immune signals to efficiently protect mosquitoes from P. berghei infection .

Experiments involving the depletion of Cactus, which usually provides protection to mosquitoes, showed no effect in the absence of APL1C, confirming that the effect of APL1C is functionally dominant in this anti-P. berghei pathway . It is hypothesized that APL1C acts as an upstream signaling node responsible for the coordinated Rel1-dependent control of multiple host protective factors .

Interaction with LRIM1 and TEP1

LRIM1 and APL1C form a disulfide-bonded complex that interacts with the complement C3-like protein TEP1, which is crucial for the mosquito's immune defense against Plasmodium parasites . The LRIM1/APL1C complex specifically binds and solubilizes a cleaved form of TEP1, lacking an intact thioester bond . This complex stabilizes mature TEP1 and promotes its binding to parasites, triggering their destruction and clearance .

Genetic Association with P. berghei Infection

Allelic haplotypes at the APL1 locus are genetically linked to the success or failure of P. berghei parasite infection .

Regulation of APL1C Protein Abundance

Plasmodium berghei infection and Cactus regulate APL1C protein abundance . APL1C protein abundance was analyzed 24 hours post-P. berghei infection in naïve, dsCactus knockdown and dsGFP control mosquitoes .

Data Tables

GeneFunctionRegulation
APL1APart of the APL1 family; function not as clearly defined as APL1CNot as well-defined as APL1C; likely part of the same regulatory mechanisms
APL1BPart of the APL1 family; function not as clearly defined as APL1CNot as well-defined as APL1C; likely part of the same regulatory mechanisms
APL1CHost defense against P. berghei; required for Rel1-mediated protectionRegulated by Rel1/Cactus pathway; Rel1 positively, Cactus negatively regulates it
LRIM1Part of the LRIM1/APL1C complex that interacts with TEP1Not available
TEP1Binds to the surface of invading parasites, triggering their destruction and clearanceNot available
ExperimentResult
Silencing of APL1CAbolishes Rel1-mediated host protection against P. berghei
Cactus depletion in the absence of APL1CNo host-protective phenotype
Treatment with dsRNA for TEP1 and LRIM1Partially reversed the dsCactus phenotype in double knockdowns, allowing development of some normal P. berghei parasites
Rel1 silencingDecreased APL1C level
Cactus silencingIncreased APL1C level
APL1C protein abundance 24 h after P. berghei infection in naïve, dsCactus knockdown and dsGFP control mosquitoesAPL1C protein abundance is regulated by P. berghei infection and Cactus.

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. Please contact 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 settle 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 default glycerol concentration is 50%, and this can serve as a reference.
Shelf Life
Shelf life depends on several factors: storage conditions, buffer composition, temperature, and the protein's inherent 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
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 manufacturing. If you require a specific tag type, please inform us; we will prioritize its implementation. *Note: Tag type is determined during production. Specify your desired tag type for preferential development.*
Synonyms
cbc; AGAP007701Protein CLP1 homolog
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
1-423
Protein Length
full length protein
Purity
>85% (SDS-PAGE)
Species
Anopheles gambiae (African malaria mosquito)
Target Names
cbc
Target Protein Sequence
MTDDKAVPRT DYKLESDSEL RFEIENKNEK VTVVLLNGQA ELFGTELVVK KPYEFVTGAK VAIFTYHGCT IELRGKPDVA YVAKETPMVM YLNANSALEH LRNKAEQEDA QGPIVMVVGP TDVGKTTLCR IFLNYAVRLG RRPIFVDLDV GQGGIAIPGT IGALLVERPA PVAEGFSQQA PLVYHYGHST PSANSTFYDV LISKLAETTL ERLQANKKAK SSGMIINTCG WVKGSGYSHI LHTVEAFEVT AIFVLDQERL YNELLRDVKG TVQVVFLPKS GGVVERTKSQ RTEARDQRIR EYFYGSKMPL FPHSFDVKFS DIKIFKVGSP PLPDSCLPLG MKAEDNYTKL VAVQPGPQLL HHILAVSFAE STDENVIQTN VAGFICVTNV NMDKQVLTVL SPQPRPLPQT ILLVSDLQFM DSH
Uniprot No.

Target Background

Function
Essential for endonucleolytic cleavage during polyadenylation-dependent pre-mRNA 3'-end formation.
Database Links
Protein Families
Clp1 family, Clp1 subfamily
Subcellular Location
Nucleus.

Q&A

What are the major immune-related protein families in Anopheles gambiae?

Anopheles gambiae possesses several important immune-related protein families that participate in defense against pathogens such as Plasmodium parasites. These include the leucine-rich repeat immune proteins (LRIMs), particularly LRIM1 and APL1C, which form complexes with thioester-containing proteins (TEPs) to activate complement-like pathways . Additionally, the CLIP-domain serine protease family plays crucial roles in melanization responses, with 110 CLIP serine proteases (cSPs) and CLIP serine protease homologs (cSPHs) annotated in the A. gambiae genome . These are divided into five subgroups (CLIPA-E) based on phylogeny, clip-domain structure, and domain arrangement . The TEP family, which includes the complement C3-like protein TEP1, is also fundamental to the mosquito's immune response .

How do LRIM1 and APL1C function in the mosquito immune system?

LRIM1 and APL1C function exclusively as a disulfide-bonded complex that circulates in the hemolymph (mosquito blood) and specifically interacts with the mature form of the complement C3-like protein TEP1 . This interaction is crucial for immune defense against Plasmodium parasites. The LRIM1/APL1C complex contains three distinct functional modules: (1) a C-terminal coiled-coil domain that serves as the binding site for TEP proteins and contributes to the specificity of complex formation, (2) a central cysteine-rich region that controls complex formation through a key intermolecular disulfide bond, and (3) an N-terminal leucine-rich repeat with a putative role in pathogen recognition . This complex is a critical component of the mosquito complement-like pathway that targets malaria parasites.

What is the role of CLIP proteins in the prophenoloxidase activation cascade?

CLIP proteins, particularly those from the CLIPB family, are core components of the prophenoloxidase (proPO) activation cascade that leads to melanization, a major immune response in insects. In Anopheles gambiae, CLIPBs are secreted into the hemolymph as zymogens and are activated sequentially by specific cleavage at the linker region between the clip and protease domains . Research has identified CLIPB9 and CLIPB10 as functional prophenoloxidase activating proteins (PAPs) that can directly cleave and activate proPO to phenoloxidase (PO) . The cascade is hierarchically organized, with CLIPCs acting upstream of CLIPBs, and the terminal conversion of proPO to active PO is mediated by PAPs, which in all insects examined belong to the CLIPB family . This activation cascade is tightly regulated and is essential for both humoral and tissue melanization responses.

What approaches should be used to study protein-protein interactions between immune-related proteins?

To study protein-protein interactions between immune-related proteins such as LRIM1, APL1C, and TEP1, researchers should employ a combination of genetic and biochemical approaches. Cell culture assays using altered key conserved structural elements can be used to assess complex formation and interaction capabilities . For instance, researchers have generated sets of LRIM1 and APL1C alleles with alterations in key conserved structural elements and assayed them in cell culture for complex formation and interaction with TEP1 .

For biochemical validation, recombinant protein production followed by in vitro interaction assays is essential. This approach has been used successfully to demonstrate the direct interaction between the LRIM1/APL1C complex and various TEP proteins . Co-immunoprecipitation assays, western blotting with specific antibodies, and SDS-PAGE analysis of SDS-stable protein complexes are valuable methods to detect physical interactions . Additionally, functional assays such as measuring phenoloxidase activity in the presence of different protein combinations can provide insights into functional interactions .

How should recombinant mosquito immune proteins be produced and purified for functional studies?

Purification protocols typically involve affinity chromatography using the added tags, followed by size exclusion chromatography to remove aggregates and obtain homogeneous protein preparations. For proteins requiring activation, such as CLIPB zymogens, controlled proteolytic activation is necessary. For instance, Factor Xa has been used to activate recombinant proCLIPB9 and proCLIPB10 in vitro . Quality control steps should include SDS-PAGE analysis, western blotting with specific antibodies, and functional assays to confirm the activity of the purified proteins before using them in further experiments.

What controls should be included when studying the activation of prophenoloxidase by CLIP proteases?

When studying the activation of prophenoloxidase (proPO) by CLIP proteases, several controls are essential to ensure the validity and specificity of the results. First, inactive precursor forms of the CLIP proteases (e.g., proCLIPB9, proCLIPB10) should be included to verify that activation is required for their function . Second, the proteolytic agent used for in vitro activation (e.g., Factor Xa) should be tested alone to ensure it does not directly activate proPO .

Additionally, researchers should include positive controls using previously validated activators of the cascade and negative controls with buffer only. Time-course experiments and dose-response curves help determine the optimal conditions for activation. When using plasma as a source of proPO (e.g., from Manduca sexta), pre-screening to ensure the absence of endogenous activation is important . Western blot analysis using anti-PO antibodies should be performed to confirm the specific cleavage of proPO to PO, alongside enzyme activity assays measuring the conversion of substrates like L-DOPA to dopachrome . Including inhibitors such as serpins can provide further evidence of the specificity of the activation process and help place the tested proteases in the activation cascade.

How can genetic variation in immune-related genes be analyzed to identify signals of adaptive evolution?

Analysis of genetic variation in immune-related genes to identify signals of adaptive evolution requires a combination of population genetics approaches. Researchers have successfully applied these methods to identify adaptive evolution in TEP1, the APL1 gene cluster, and LRIM1 in Anopheles gambiae populations . The process involves sequencing these genes in multiple natural populations and applying several statistical tests to detect departures from neutral evolution.

Key analysis methods include: (1) detecting enrichment of high-frequency-derived alleles using normalized Fay and Wu's H statistic (Hnorm), (2) measuring increases in haplotype homozygosity using the Ewens-Watterson statistic (EW), and (3) combining these statistics into a composite statistic (HEW) with appropriate correction for multiple testing . For instance, researchers found evidence for positive selection at the TEP1 locus (HEW corrected p=0.0233) and at LRIM1 (HEW corrected p=0.0336) in the M-form population of A. gambiae . When analyzing paralogous genes like the APL1 family, each paralog should be analyzed separately to identify which specific member shows the strongest signal of selection . The relative strength of selection on different members of a protein complex can also be assessed by comparing their positions as outliers in statistical distributions .

How can contradictory experimental results in mosquito immunity research be reconciled?

Contradictory experimental results in mosquito immunity research can arise from differences in experimental design, mosquito strains, parasite species, or environmental conditions. To reconcile such contradictions, researchers should first carefully compare methodology across studies, noting differences in reagents, protocols, and genetic backgrounds of model organisms.

A systematic approach includes: (1) Replicating key experiments using standardized protocols across laboratories, (2) Using multiple complementary techniques to address the same question (e.g., combining genetic knockdowns with biochemical assays), (3) Testing hypotheses across different mosquito strains and parasite isolates, and (4) Employing more sophisticated statistical analyses that can account for variability in experimental conditions.

When contradictions arise in the specific roles of proteins like CLIPB10, researchers can perform careful structure-function analyses using recombinant proteins with site-directed mutations , or create genetic knockouts using CRISPR-Cas9 rather than relying solely on RNAi-based knockdowns. Additionally, comprehensive testing of protein interactions in vitro and in vivo can help distinguish direct from indirect effects. For example, the study showing that CLIPB10 can directly activate prophenoloxidase helped clarify its role in the melanization cascade, despite previous uncertainty about its position in the pathway .

What statistics should be used when analyzing phenotypic effects of immune protein knockdowns?

For survival experiments, Kaplan-Meier survival analysis with log-rank tests is appropriate for comparing different experimental groups. When measuring parasite loads or enzyme activities (e.g., PO activity), data typically follow normal distributions after appropriate transformations (log-transformation is common), allowing for parametric tests such as t-tests or ANOVA with appropriate post-hoc tests for multiple comparisons.

It's important to control for batch effects and biological variability by including appropriate controls in each experimental batch and using mixed-effects models when analyzing data pooled across multiple experiments. For protein-protein interaction studies, quantification of band intensities from western blots should be normalized to appropriate loading controls, and statistical comparisons should account for technical variability across replicates. In all cases, reporting of p-values should be accompanied by effect sizes and confidence intervals to provide a complete picture of the biological significance of the findings.

How can the functional redundancy in the mosquito immune system be experimentally addressed?

Addressing functional redundancy in the mosquito immune system, such as that observed between CLIPB9 and CLIPB10 in the prophenoloxidase activation cascade , requires sophisticated experimental approaches. A comprehensive strategy should include:

  • Simultaneous knockdown or knockout of multiple redundant genes using combinatorial RNAi or CRISPR-Cas9 multiplexing, comparing the phenotypes to single gene perturbations

  • Complementation experiments where one knocked-down gene is rescued by expression of its paralog

  • Domain-swapping experiments between paralogs to identify which protein regions confer specific functions versus shared functions

  • Quantitative biochemical assays to compare the enzymatic efficiency of redundant proteins (e.g., kinetic analysis of proPO activation by CLIPB9 versus CLIPB10)

  • Temporal and spatial expression analysis using qRT-PCR and in situ hybridization to determine if redundant proteins are expressed in different tissues or developmental stages

This multi-faceted approach can reveal whether apparent redundancy serves as functional backup or if the paralogs have subtle specializations that aren't immediately obvious in standard assays. For example, while both CLIPB9 and CLIPB10 can function as PAPs, differences in their regulation or enzymatic properties may make them more important in different physiological contexts, such as tissue versus humoral melanization .

What are the molecular mechanisms of regulation in the mosquito complement-like pathway?

The regulation of the mosquito complement-like pathway involves complex molecular mechanisms that balance immune activation with prevention of harmful excessive responses. A central regulatory mechanism involves the interaction between clip-domain serine proteases and their inhibitors, the serpins. Specifically, SRPN2 has been identified as a key inhibitor of the proPO activation cascade in A. gambiae .

Biochemical studies have shown that SRPN2 forms SDS-stable protein complexes with activated CLIPB10, effectively inhibiting its activity in vitro at a stoichiometry of 1.89:1 . This inhibition is highly specific and represents a crucial regulatory checkpoint in the cascade. Additionally, the pathway is regulated through the controlled proteolytic activation of the clips themselves, with evidence suggesting a hierarchical organization where CLIPCs act upstream of CLIPBs .

At the protein complex level, the LRIM1/APL1C complex specifically interacts with mature forms of TEP proteins through the coiled-coil domain . This interaction likely stabilizes the activated TEP proteins and prevents their inappropriate activation or degradation. The formation of the LRIM1/APL1C complex itself is regulated through specific disulfide bonding involving key cysteine residues in the central cysteine-rich region .

Regulatory mechanisms may also include feedback loops and cross-talk between different immune pathways, although these aspects remain less well understood in mosquitoes compared to model organisms like Drosophila.

How can structural biology approaches advance our understanding of mosquito immune protein complexes?

Structural biology approaches offer powerful tools for understanding the molecular basis of mosquito immune protein complexes and their functions. To advance research in this area, scientists could employ:

  • X-ray crystallography to determine high-resolution structures of key proteins (e.g., LRIM1/APL1C complex, activated CLIPB10) and their interactions with binding partners

  • Cryo-electron microscopy for larger complexes, such as the full TEP1/LRIM1/APL1C assembly

  • NMR spectroscopy to study the dynamics of protein-protein interactions and conformational changes upon activation

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS) to map interaction interfaces and conformational changes

  • Small-angle X-ray scattering (SAXS) for lower-resolution envelope structures of complexes in solution

These approaches could reveal the structural basis for the three distinct modules identified in LRIM1 and APL1C: the C-terminal coiled-coil domain that binds TEP proteins, the central cysteine-rich region controlling complex formation, and the N-terminal leucine-rich repeat with potential pathogen recognition function . Understanding the structural basis of these interactions could enable rational design of molecules to modulate mosquito immunity against malaria parasites.

For CLIP proteases, structural studies could elucidate the molecular basis of zymogen activation, substrate specificity, and inhibition by serpins like SRPN2. This information would be valuable for placing different CLIPs in their correct hierarchical positions in the activation cascades and understanding how they achieve specificity despite their sequence similarity.

What are the optimal RNAi protocols for knockdown of immune genes in Anopheles gambiae?

Effective RNAi-based knockdown of immune genes in Anopheles gambiae requires optimization of several parameters. Based on established protocols, researchers should:

  • Design dsRNA fragments of 300-600 bp in length, targeting unique regions of the gene of interest to minimize off-target effects

  • Include appropriate controls, such as dsRNA targeting GFP or LacZ genes that are absent from the mosquito genome

  • Inject young adult female mosquitoes (1-2 days post-emergence) with 69 nl of dsRNA solution (3 μg/μl) into the thorax

  • Maintain injected mosquitoes under standard insectary conditions (27°C, 80% humidity, 12:12 light:dark cycle)

  • Allow 3-4 days for gene silencing to take effect before challenging with pathogens or conducting assays

  • Verify knockdown efficiency by qRT-PCR, with a target reduction of at least 70% in mRNA levels

For genes involved in melanization pathways, such as CLIPB10, knockdown efficiency can be functionally validated by assessing melanization responses to Sephadex beads or Plasmodium parasites . The timing of knockdown relative to immune challenge is critical, as different genes may have different temporal expression patterns following immune stimulation. Additionally, some genes may have maternal effects or roles in multiple tissues, requiring careful experimental design to distinguish these different functions.

How can recombinant CLIP proteases be activated in vitro for functional studies?

In vitro activation of recombinant CLIP proteases is a critical step for functional studies of these zymogens. The protocol typically involves:

  • Expression of the zymogen form (e.g., proCLIPB10) with appropriate tags (His-tag) in expression systems like E. coli

  • Purification using affinity chromatography (Ni-NTA for His-tagged proteins) followed by size exclusion chromatography

  • Incorporation of an exogenous protease cleavage site (e.g., Factor Xa) between the tag and the protein of interest

  • Controlled proteolytic activation using the specific protease (e.g., Factor Xa) under optimized conditions (typically 4-16 hours at room temperature)

  • Verification of activation by SDS-PAGE and western blotting, looking for the appearance of the characteristic cleaved form

  • Purification of the activated protease from the activation mix if necessary

This approach has been successfully employed for CLIPB9 and CLIPB10, enabling studies of their activity as PAPs . The activated proteases can then be used in functional assays, such as PO activation assays using purified proPO or hemolymph from model insects like Manduca sexta. It's important to include controls such as unactivated zymogens and the activation protease alone to ensure that the observed effects are specifically due to the activated CLIP protease .

What methods can be used to study the interaction between recombinant serpins and CLIP proteases?

Studying the interaction between recombinant serpins (such as SRPN2) and CLIP proteases (such as CLIPB10) requires specific biochemical approaches that can detect both physical interactions and functional inhibition. Recommended methods include:

  • SDS-stable complex formation assay: Mix purified recombinant serpin with activated CLIP protease at different molar ratios, then analyze by SDS-PAGE and western blotting. The formation of high-molecular-weight bands that are resistant to SDS denaturation is indicative of the covalent complex formed between the serpin and its target protease .

  • Determination of stoichiometry of inhibition (SI): Incubate increasing concentrations of serpin with a fixed amount of active protease, then measure the residual protease activity using appropriate substrates. The SI value (the molar ratio of serpin to protease at which protease activity is completely inhibited) provides information about the efficiency of inhibition. For example, SRPN2 inhibits CLIPB10 at a stoichiometry of 1.89:1 .

  • Enzyme kinetics: Measure the rate of substrate cleavage by the protease in the presence of varying concentrations of serpin to determine inhibition constants (Ki) and the mode of inhibition (competitive, non-competitive, or uncompetitive).

  • Functional rescue experiments: In mosquitoes where the serpin has been silenced, resulting in constitutive melanization, co-silencing of the target CLIP protease should reverse the phenotype if the protease is a physiological target of the serpin. This approach has been used to show that CLIPB10 knockdown partially reversed melanotic tumor formation induced by Serpin 2 silencing .

These methods provide complementary information about the physical interaction between serpins and CLIP proteases and the functional consequences of these interactions in regulating the proteolytic cascades involved in mosquito immunity.

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.