CFB Human

Complement Factor B Human Recombinant
Shipped with Ice Packs
In Stock

Description

Biological Functions

CFB drives the alternative complement pathway through two mechanisms:

  1. C3 Convertase Formation: Cleaved by Factor D into fragments Ba (33 kDa) and Bb (60 kDa). Bb binds C3b to form C3 convertase (C3bBb), amplifying C3 cleavage .

  2. C5 Convertase Formation: Bb combines with C3b to cleave C5, initiating membrane attack complex (MAC) assembly .

Additional Roles:

  • Regulates preactivated B-lymphocyte proliferation (Bb promotes; Ba inhibits) .

  • Modulates adipose tissue metabolism and systemic inflammation .

Diagnostic Methods:

  • ELISA: Quantifies CFB levels (sensitivity: 0.1–10 µg/ml) .

  • Hemolytic Assays: Measures alternative pathway activity (AH50 test) .

  • Genetic Testing: Identifies pathogenic variants (e.g., p.Ser367Arg in aHUS) .

Therapeutic Targets:

  • Iptacopan: Oral CFB inhibitor reducing hemolysis in PNH (hemoglobin improvement: +2.4 g/dL) .

  • siRNA Therapies: Suppress synovial inflammation in rheumatoid arthritis models .

Production and Stability

  • Recombinant CFB: Purified via His-Tag chromatography; stable at 4°C for 2–4 weeks .

  • Native CFB: Isolated from human plasma; requires -20°C storage with carrier proteins (e.g., 0.1% HSA) .

Research Advancements

  • GWAS Links: SNPs near CFB associate with visceral fat, triglycerides, and hypertension (e.g., rs12614) .

  • Gene Expression: Adipose tissue CFB overexpression drives insulin resistance in metabolic syndrome models .

  • Pathway Analysis: Low CFB in LUAD downregulates oxidative phosphorylation and complement cascades (NES = -1.82; FDR <0.05) .

Clinical Implications

CFB’s dual role as a complement effector and metabolic regulator positions it as a biomarker for cancer prognosis and a target for immune-mediated diseases. Ongoing trials targeting CFB (e.g., iptacopan in IgA nephropathy) highlight its therapeutic potential . Future studies must address CFB’s tissue-specific roles and interaction with inflammatory cytokines like IL-6 and TNFα .

Product Specs

Introduction
Complement Factor B, also known as CFB, is a protein involved in the complement system, a part of the immune system. It helps activate the alternative pathway of complement activation. This pathway is important for destroying pathogens (harmful microorganisms) and removing damaged cells from the body. When activated, CFB is cleaved into two parts: Ba and Bb. Bb is the active part and works with another protein, C3b, to form a complex that helps break down pathogens.
Description
This product is a laboratory-made version of the human CFB protein. It is produced in bacteria (E.coli) and purified to a high degree. It is provided as a solution with a concentration of 1 mg/ml. This recombinant protein is a single chain of 528 amino acids with a molecular weight of 59.4 kDa. It contains a His-tag at the N-terminus for easy purification.
Physical Appearance
Clear and colorless solution that has been sterilized by filtration.
Formulation
This solution contains 1 mg of CFB per ml, 20mM Tris-HCl buffer (pH 8.0), 0.4M Urea, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the solution refrigerated at 4°C. For longer storage, freeze the solution at -20°C. To ensure stability during long-term storage, it is advised to add a carrier protein like HSA or BSA (0.1%). Avoid repeatedly freezing and thawing the solution.
Purity
The purity of this product is over 85%, as determined by SDS-PAGE, a method used to separate and analyze proteins.
Synonyms
Complement factor B (EC:3.4.21.47), C3/C5 convertase, Glycine-rich beta glycoprotein, GBG, PBF2, Properdin factor B, Complement factor B Ba fragment, Complement factor B Bb fragment, CFB, Complement Factor B, BFD, AHUS4, BF, BFD, CFAB, FB, FBI12, H2-Bf.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSKIVLDPS GSMNIYLVLD GSDSIGASNF TGAKKCLVNL IEKVASYGVK PRYGLVTYAT YPKIWVKVSE ADSSNADWVT KQLNEINYED HKLKSGTNTK KALQAVYSMM SWPDDVPPEG WNRTRHVIIL MTDGLHNMGG DPITVIDEIR DLLYIGKDRK NPREDYLDVY VFGVGPLVNQ VNINALASKK DNEQHVFKVK DMENLEDVFY QMIDESQSLS LCGMVWEHRK GTDYHKQPWQ AKISVIRPSK GHESCMGAVV SEYFVLTAAH CFTVDDKEHS IKVSVGGEKR DLEIEVVLFH PNYNINGKKE AGIPEFYDYD VALIKLKNKL KYGQTIRPIC LPCTEGTTRA LRLPPTTTCQ QQKEELLPAQ DIKALFVSEE EKKLTRKEVY IKNGDKKGSC ERDAQYAPGY DKVKDISEVV TPRFLCTGGV SPYADPNTCR GDSGGPLIVH KRSRFIQVGV ISWGVVDVCK NQKRQKQVPA HARDFHINLF QVLPWLKEKL QDEDLGFL.

Q&A

What is Complement Factor B and what physiological roles does it play in humans?

Complement Factor B (CFB) is a serum protein that functions as a key component of the alternative pathway in the complement system. As part of the innate immune response, CFB acts as a zymogen (inactive enzyme precursor) that, when cleaved by Factor D, forms the C3 convertase (C3bBb) complex. This complex is central to the amplification phase of the complement cascade.

In physiological conditions, CFB contributes to:

  • Pathogen clearance through the alternative complement pathway

  • Immune surveillance and homeostasis

  • Removal of damaged cells and cellular debris

  • Modulation of inflammatory responses

Recent research has also identified CFB as a determinant of metabolic health, with studies showing it influences adipose tissue distribution, glucose homeostasis, blood pressure regulation, and left ventricular mass in experimental models . CFB is elevated in adipose tissue and serum from patients with type 2 diabetes mellitus and cardiovascular disease, suggesting broader physiological implications beyond traditional immune functions .

How does genetic variation in the CFB locus affect human health outcomes?

Genetic variation at the human CFB locus has been associated with several important health outcomes through both direct functional effects and altered expression levels. Analysis of the human CFB locus has revealed significant associations between specific genetic variants and disease phenotypes:

  • Three single-nucleotide polymorphisms (SNPs) at the CFB locus have been significantly associated with visceral adiposity, hypertension, and CFB gene expression levels .

  • In silico analysis identified two cis-regulated expression quantitative trait loci (eQTLs) for CFB expression that are significantly associated with visceral fat accumulation, circulating triglyceride levels, and hypertension in genome-wide association studies (GWAS) .

These genetic associations support a causal role for CFB in metabolic and cardiovascular conditions rather than it being merely a biomarker. The identification of specific risk-associated genetic variants provides a foundation for stratifying patients and developing targeted therapeutic approaches for conditions involving CFB dysregulation.

What are the canonical functional activities researchers measure when studying CFB?

When investigating CFB in research settings, scientists typically assess four key functional activities that reflect its biological roles:

Functional ActivityMeasurement MethodBiological Significance
C3b BindingSurface Plasmon Resonance (SPR)Determines affinity for forming the initial C3bB complex
Decay-Accelerating Activity (DAA)Surface Plasmon Resonance (SPR)Measures influence on C3 convertase stability
Cofactor Activity (CA)Fluorescence assays with 8-anilinonaphthalene-1-sulfonic acid (ANS)Assesses ability to serve as cofactor for Factor I-mediated cleavage
Cell ProtectionHemolysis assaysEvaluates capacity to protect cells from complement-mediated lysis

These assays provide complementary information about different aspects of CFB function. For example, in studies comparing GEM103 (a recombinant human Complement Factor H) with native human CFH, researchers used these methods to demonstrate functional equivalence between the recombinant and native forms, confirming that the recombinant protein maintains normal complement-regulating capabilities .

What are the optimal methods for measuring CFB activity in human samples?

When designing experiments to measure CFB activity in human samples, researchers should consider multiple methodological approaches to ensure comprehensive and reliable assessment:

Surface Plasmon Resonance (SPR) Analysis:

  • Platform recommendation: BiaCore T200 (GE Healthcare) or equivalent systems

  • Application: Measures real-time protein-protein interactions, allowing quantification of:

    • Binding affinity between CFB and C3b

    • Formation kinetics of the C3bB complex

    • Decay-accelerating activity of CFB-containing complexes

  • Advantages: Provides label-free, real-time kinetic data with high sensitivity

Functional Enzyme Assays:

  • Cofactor activity measurement using fluorescent indicators such as 8-anilinonaphthalene-1-sulfonic acid (ANS)

  • Equipment: Fluorescence plate readers (e.g., SpectraMax M5e or equivalent)

  • Protocol considerations: Standardization of reagent concentrations, incubation times, and temperature is critical for reproducibility

Cell-Based Functional Tests:

  • Hemolysis assays to evaluate complement-mediated lysis and protective effects

  • Controls must include both positive (maximum lysis) and negative (no complement) conditions

  • Data analysis should include dose-response curves rather than single-point measurements

The selection of appropriate methods depends on the specific research question, available equipment, and type of samples. For clinical samples, which may have limited volume or complex matrices, optimized protocols that minimize sample requirements while maximizing sensitivity should be developed .

How should researchers distinguish between native and recombinant forms of CFB in experimental settings?

Distinguishing between native (serum-derived) and recombinant forms of CFB requires a multi-faceted characterization approach:

Comparative Functional Analysis:

  • Side-by-side testing of native serum-derived CFB (sdCFB) and recombinant CFB

  • Evaluation across multiple functional parameters:

    • C3b binding affinity (using SPR)

    • C3 convertase formation rates

    • Enzymatic activities

    • Cellular protection capabilities

  • Analysis of dose-response relationships to identify potency differences

Structural Characterization:

  • Mass spectrometry to:

    • Confirm protein mass

    • Identify post-translational modifications

    • Detect structural variations

  • Glycosylation analysis to characterize differences in carbohydrate patterns

  • Circular dichroism spectroscopy to compare secondary structure elements

Quality Control Metrics:

  • Purity assessment via SDS-PAGE with silver staining

  • Western blotting with specific antibodies to confirm identity

  • Size-exclusion chromatography to detect aggregates or oligomeric forms

In research comparing GEM103 (recombinant human CFH) with native CFH, investigators employed multiple functional assays to validate that the recombinant protein maintained equivalent biological activity, demonstrating the importance of comprehensive characterization when working with recombinant complement factors .

What considerations are important when designing genetic studies of CFB in human populations?

When designing genetic studies investigating CFB in human populations, researchers should address several critical considerations:

Study Population Selection:

  • Define clear inclusion/exclusion criteria based on phenotypes of interest

  • Consider population stratification and genetic ancestry

  • Evaluate the need for replication cohorts with different demographic characteristics

Genetic Variation Coverage:

  • Target both common and rare variants at the CFB locus

  • Include regulatory regions that may affect expression (promoters, enhancers, etc.)

  • Consider linkage disequilibrium patterns when selecting tag SNPs

Phenotype Characterization:

  • Collect comprehensive phenotypic data related to complement activation

  • Include relevant metabolic and cardiovascular measurements

  • Standardize phenotyping methods across study sites

Analytical Approaches:

  • Conduct expression quantitative trait loci (eQTL) analysis to identify variants affecting CFB expression

  • Perform Mendelian randomization to assess causality

  • Consider gene-environment interactions that may modify genetic effects

Integration with Functional Data:

  • Connect genetic findings with functional studies to validate biological mechanisms

  • Assess how identified variants affect protein structure or function

  • Correlate genotypes with complement activation biomarkers

Research has identified significant associations between CFB genetic variants and cardiometabolic traits, including visceral adiposity, hypertension, and circulating triglycerides . These findings demonstrate the value of well-designed genetic studies in uncovering the role of CFB in human disease.

How does CFB contribute to metabolic syndrome pathophysiology?

CFB plays a multifaceted role in metabolic syndrome pathophysiology, with evidence from both animal models and human studies suggesting mechanistic connections between CFB and multiple components of this condition:

Adipose Tissue Distribution and Function:

  • CFB influences adipose tissue distribution, with genetic deletion of Cfb in spontaneously hypertensive rats resulting in altered fat deposition patterns

  • The protein appears to affect adipocyte metabolic functions and inflammatory state

  • Human genetic studies have linked CFB variants to visceral adiposity

Glucose Homeostasis:

  • Cfb deletion improves glucose tolerance and insulin sensitivity in experimental models

  • CFB may influence insulin signaling pathways in target tissues

  • The protein potentially contributes to pancreatic β-cell dysfunction in diabetic conditions

Blood Pressure Regulation:

  • CFB directly or indirectly drives novel mechanisms for hypertension

  • Deletion of the Cfb gene lowers blood pressure in spontaneously hypertensive rats

  • Human SNPs at the CFB locus are associated with hypertension in genome-wide association studies

Cardiac Remodeling:

  • CFB influences left ventricular mass and cardiac hypertrophy

  • Cfb deletion protects against cardiac stress and improves fractional shortening

  • These effects appear to operate through both blood pressure-dependent and independent mechanisms

The widespread amelioration of metabolic and cardiovascular phenotypes through CFB deletion in experimental models suggests this complement factor may represent a novel therapeutic target for metabolic syndrome. Human genetic evidence supports the translational relevance of these findings, with SNPs at the CFB locus associated with related traits in genome-wide studies .

What methodological approaches are most effective for investigating CFB's role in age-related macular degeneration?

Age-related macular degeneration (AMD) research involving CFB requires specialized methodological approaches tailored to this ocular disorder:

Genetic Analysis Techniques:

  • Comprehensive genotyping of CFB and related complement genes (CFH, C3, etc.)

  • Risk haplotype identification and association testing

  • Analysis of genetic interactions between CFB and CFH variants

Functional Complement Assays:

  • Measurement of CFB levels in serum and ocular fluids

  • Assessment of complement activation products in patient samples

  • Evaluation of CFB's interaction with AMD-associated variants of CFH

Therapeutic Development Methods:

  • Recombinant protein production approaches (as demonstrated with GEM103)

  • Functional validation comparing recombinant versus native proteins

  • In vitro assays measuring C3b binding, decay-accelerating activity, and cofactor activity using surface plasmon resonance and other techniques

Translational Models:

  • Cell culture systems using retinal pigment epithelial cells

  • Ex vivo choroidal explant cultures

  • Animal models of complement-mediated retinal degeneration

Clinical Investigation Approaches:

  • Patient stratification based on genetic complement risk profiles

  • Biomarker evaluation in different stages of AMD

  • Design of intervention studies targeting specific complement components

Recent research has focused on developing recombinant complement factors as potential AMD treatments. For example, GEM103, a recombinantly produced full-length version of human Complement Factor H (CFH), is under clinical investigation for AMD treatment in individuals carrying risk-associated genetic variants of CFH. Functional studies have confirmed that GEM103 shows similar in vitro activity to native CFH across multiple complement-related assays .

How do research methodologies differ when studying CFB in cardiovascular versus immunological contexts?

Studying CFB across cardiovascular and immunological contexts requires distinct yet complementary methodological approaches, reflecting the diverse roles of this protein:

Research AspectCardiovascular ContextImmunological Context
Sample TypesCardiac tissue, vascular cells, plasmaImmune cells, serum, inflammatory exudates
Functional Assays- Blood pressure measurements
- Cardiac imaging (echocardiography, MRI)
- Vascular function tests
- Complement activation assays
- Pathogen clearance evaluation
- Inflammatory mediator production
Key Endpoints- Left ventricular mass
- Blood pressure
- Vascular resistance
- Cardiac remodeling
- C3 convertase formation
- Opsonization efficiency
- Cell lysis
- Inflammatory cytokine patterns
Genetic Approaches- Focus on cardiometabolic trait associations
- Blood pressure-related SNPs
- Focus on infectious/autoimmune disease associations
- Complement deficiency consequences
Model Systems- Spontaneously hypertensive rats
- Pressure-overload models
- Metabolic syndrome models
- Complement activation assays
- Infection models
- Autoimmune disease models

When designing studies, researchers should consider these contextual differences while also recognizing the interconnected nature of cardiovascular and immunological processes. For example, in spontaneously hypertensive rats, Cfb deletion improved both metabolic parameters and cardiovascular outcomes, demonstrating how CFB functions at the intersection of these systems .

Integration of complementary methodologies can provide a more comprehensive understanding of CFB's diverse biological roles and identify context-specific therapeutic opportunities.

How should researchers address data inconsistencies when analyzing CFB's functional effects across different experimental systems?

When confronted with inconsistent data regarding CFB's functional effects across experimental systems, researchers should implement a systematic analytical framework:

Root Cause Analysis:

  • Evaluate methodological differences that might explain discrepancies:

    • Assay conditions (pH, ionic strength, temperature)

    • Protein source and quality (recombinant vs. native)

    • Cell types or animal models used

  • Assess biological variables that could influence outcomes:

    • Genetic background differences

    • Age and sex effects

    • Disease state or model characteristics

Statistical Resolution Approaches:

  • Perform sensitivity analyses to identify threshold conditions affecting outcomes

  • Use meta-analytical techniques to synthesize results across multiple studies

  • Implement Bayesian modeling to incorporate prior knowledge and uncertainty

Experimental Reconciliation Strategies:

  • Design side-by-side comparison studies under standardized conditions

  • Develop dose-response curves rather than relying on single concentrations

  • Test for interaction effects with other complement components

Conceptual Framework Development:

  • Consider that apparent contradictions may reflect context-dependent roles

  • Develop testable hypotheses that could explain seemingly discrepant results

  • Design critical experiments specifically to resolve key contradictions

When investigating GEM103 (recombinant CFH), researchers employed multiple independent functional assays to provide robust validation of its activity compared to native CFH, demonstrating the value of comprehensive, multi-assay approaches in addressing potential data inconsistencies .

What statistical approaches are recommended for analyzing associations between CFB genetic variants and disease phenotypes?

Analyzing associations between CFB genetic variants and disease phenotypes requires sophisticated statistical approaches to ensure robust, reproducible, and biologically meaningful results:

Primary Association Testing:

  • Linear or logistic regression models adjusted for relevant covariates

  • Inclusion of population stratification controls (e.g., principal components)

  • Appropriate multiple testing correction (Bonferroni, false discovery rate)

Extended Genetic Analysis:

  • Haplotype-based association testing to capture effects of variant combinations

  • Conditional analysis to identify independent signals at the CFB locus

  • Gene-gene interaction testing, particularly with other complement genes

Causality Assessment:

  • Mendelian randomization to infer causality from genetic associations

  • Mediation analysis to identify pathways linking CFB to disease outcomes

  • Polygenic risk score development incorporating CFB and related variants

Integration with Expression Data:

  • Expression quantitative trait loci (eQTL) analysis to link variants to CFB expression

  • Joint analysis of genomic and transcriptomic data using methods like PrediXcan

  • Network analysis incorporating protein-protein interactions

Translational Statistics:

  • Risk prediction modeling to evaluate clinical utility

  • Number needed to genotype calculations for potential interventions

  • Development of genetic risk scores with CFB variants

In research on CFB's role in metabolic syndrome, researchers identified two cis-regulated eQTLs for CFB expression significantly associated with visceral fat, circulating triglycerides, and hypertension in genome-wide association studies . These findings demonstrate the value of integrating genetic association and expression data to understand CFB's role in human disease.

How can researchers effectively compare results from animal models of CFB function with human clinical data?

Bridging the gap between animal model findings and human clinical data on CFB function requires systematic comparative approaches:

Comparative Genomics Framework:

  • Sequence alignment analysis of CFB across species

  • Identification of conserved versus divergent functional domains

  • Assessment of species-specific regulatory elements affecting CFB expression

Cross-Species Experimental Design:

  • Parallel study designs in animal models and human samples

  • Standardized outcome measures across species

  • Use of humanized animal models expressing human CFB variants

Translational Biomarker Strategy:

  • Identification of conserved biomarkers that reflect CFB activity across species

  • Validation of these markers in both experimental models and human samples

  • Development of assays applicable to both animal and human specimens

Integrative Data Analysis:

  • Meta-regression incorporating both animal and human studies

  • Bayesian hierarchical modeling to account for species differences

  • Network analysis to identify conserved pathways versus species-specific mechanisms

Predictive Modeling:

  • Development of in silico models to predict human responses based on animal data

  • Calibration of these models using available human data

  • Sensitivity analysis to identify key parameters driving species differences

Research on CFB in spontaneously hypertensive rats demonstrated widespread amelioration of metabolic and cardiovascular phenotypes through CFB deletion. In parallel, human genetic studies identified CFB variants associated with similar phenotypes, suggesting conserved mechanisms across species . This complementary evidence strengthens the translational potential of targeting CFB for human metabolic and cardiovascular diseases.

What novel methodologies are emerging for studying CFB's tissue-specific functions?

Several innovative methodologies are being developed to investigate CFB's tissue-specific functions with unprecedented precision and depth:

Single-Cell Genomics Approaches:

  • Single-cell RNA sequencing to map CFB expression across tissue-resident cell populations

  • Spatial transcriptomics to visualize CFB expression patterns within tissue architecture

  • Single-cell ATAC-seq to identify cell-type-specific regulatory elements controlling CFB expression

Advanced Imaging Techniques:

  • Intravital microscopy with fluorescently tagged CFB to track dynamics in living tissues

  • Super-resolution microscopy to visualize CFB-protein interactions at nanoscale resolution

  • Label-free imaging methods to detect endogenous CFB activity in intact tissues

Tissue-Specific Genetic Manipulation:

  • Conditional and inducible knockout systems targeting CFB in specific tissues

  • CRISPR-Cas9-based approaches for precise genetic editing

  • AAV-mediated gene delivery for tissue-targeted CFB modulation

Ex Vivo Tissue Systems:

  • Organ-on-chip technologies incorporating tissue-specific microenvironments

  • Patient-derived organoids to model CFB function in human tissues

  • Perfused tissue explants maintaining physiological complement activation

Multi-omics Integration:

  • Combined proteomics, metabolomics, and transcriptomics to characterize tissue-specific CFB networks

  • Computational models integrating multiple data types to predict tissue-specific effects

  • Systems biology approaches to understand CFB in the context of tissue-specific immune environments

These emerging methodologies promise to advance our understanding of how CFB functions across different tissues, potentially revealing new therapeutic opportunities for conditions ranging from age-related macular degeneration to metabolic syndrome and cardiovascular disease.

What are the key research gaps in understanding CFB's role in inflammatory diseases?

Despite significant advances, several critical knowledge gaps remain in understanding CFB's role in inflammatory diseases:

Mechanistic Understanding:

  • Precise molecular mechanisms by which CFB contributes to tissue-specific inflammation

  • Upstream regulators controlling CFB expression during different inflammatory states

  • Non-canonical functions of CFB beyond the alternative complement pathway

  • Cross-talk between CFB and other inflammatory mediators

Temporal Dynamics:

  • CFB's role in the initiation versus propagation of inflammatory responses

  • Temporal changes in CFB function during acute versus chronic inflammation

  • Resolution phase dynamics and CFB's potential role in inflammation resolution

Genetic and Environmental Interactions:

  • How genetic variants in CFB modify inflammatory disease risk and progression

  • Environmental factors that influence CFB expression and function

  • Epigenetic regulation of CFB during inflammatory states

Therapeutic Targeting Challenges:

  • Optimal strategies for tissue-specific inhibition of pathological CFB activity

  • Balancing beneficial versus harmful effects of CFB modulation

  • Biomarkers to identify patients likely to benefit from CFB-targeted therapies

Translational Validation:

  • Validation of animal model findings in human inflammatory conditions

  • Establishment of relevant human experimental systems

  • Development of standardized assays for clinical application

Addressing these research gaps will require interdisciplinary approaches combining immunology, genetics, biochemistry, and clinical research. Understanding CFB's nuanced roles in inflammation could lead to novel therapeutic strategies for conditions ranging from autoimmune disorders to metabolic inflammation and age-related diseases.

What therapeutic approaches targeting CFB are under development and how are they being evaluated?

Several innovative therapeutic approaches targeting CFB are in various stages of development, each with specific evaluation methodologies:

Recombinant Protein Therapies:

  • GEM103: A recombinantly produced full-length version of human Complement Factor H (CFH) under clinical investigation for age-related macular degeneration

  • Evaluation methods:

    • In vitro functional assays measuring C3b binding, decay-accelerating activity

    • Cofactor activity assessment using purified complement components

    • Cell protection assays in activated human serum

Small Molecule Inhibitors:

  • Compounds targeting CFB enzymatic activity or protein-protein interactions

  • Evaluation approaches:

    • High-throughput screening against recombinant CFB

    • Structure-based drug design using CFB crystal structures

    • In vitro and ex vivo functional assays in human samples

Antibody-Based Therapeutics:

  • Monoclonal antibodies and antibody fragments targeting CFB

  • Testing methodologies:

    • Binding affinity and specificity characterization

    • Functional neutralization in complement activation assays

    • Pharmacokinetic/pharmacodynamic studies in animal models

RNA-Based Therapeutics:

  • Antisense oligonucleotides and siRNA approaches to reduce CFB expression

  • Evaluation strategies:

    • Target engagement and knockdown efficiency assessment

    • Tissue-specific delivery optimization

    • Off-target effect analysis

Preclinical to Clinical Translation:

  • Animal models of CFB-related diseases (metabolic syndrome, AMD)

  • Biomarker development to monitor CFB activity in clinical samples

  • Patient stratification strategies based on CFB genetic profiles

The development of compounds targeting CFB, combined with evidence of its role in multiple disease processes, indicates significant therapeutic potential. Research suggests CFB could be a valid therapeutic target for treating or preventing progression of metabolic syndrome and related conditions .

What are the most important considerations for designing comprehensive CFB research programs?

Designing comprehensive CFB research programs requires strategic integration of multiple approaches to address the complex biology of this complement factor. Research programs should incorporate:

  • Multi-level investigation spanning molecular mechanisms to clinical outcomes

  • Complementary methodologies including genetic, biochemical, and functional approaches

  • Cross-disciplinary collaboration between immunologists, geneticists, and clinical researchers

  • Translational focus connecting basic discoveries to therapeutic applications

  • Validation across multiple model systems and human samples

The discovery that CFB functions as a determinant of both metabolic and cardiovascular traits highlights the importance of investigating this factor across traditionally separate research domains. Future research programs should consider these diverse roles when designing experiments and interpreting results .

How might findings from CFB research transform our understanding of human disease mechanisms?

CFB research has potential to transform our understanding of disease mechanisms by revealing unexpected connections between complement activation and conditions not traditionally associated with immunity. The identification of CFB as a link between inflammation, metabolism, and cardiovascular health challenges conventional paradigms and suggests new integrative models of disease pathogenesis.

The development of GEM103, a recombinant human complement factor H, demonstrates how basic research on complement proteins can lead to novel therapeutic approaches for complex conditions like age-related macular degeneration . Similarly, the discovery of CFB's role in metabolic syndrome opens new avenues for intervention in this increasingly prevalent condition .

Product Science Overview

Structure and Function

Complement Factor B circulates in the blood as a single-chain polypeptide. Upon activation of the alternative pathway, it is cleaved by complement factor D into two fragments: Ba and Bb . The Ba fragment is non-catalytic, while the Bb fragment is a serine protease that associates with C3b to form the alternative pathway C3 convertase . This convertase is essential for the amplification of the complement response, leading to the opsonization of pathogens, recruitment of inflammatory cells, and lysis of target cells.

Genetic and Molecular Characteristics

Complement Factor B is encoded by the CFB gene, which is located in the major histocompatibility complex (MHC) class III region on chromosome 6 . The protein has a molecular weight of approximately 84.1 kDa, but it can appear as 90-120 kDa on SDS-PAGE due to glycosylation .

Recombinant Production

Recombinant human Complement Factor B is produced using various expression systems, including HEK293 cells and Escherichia coli . The recombinant protein is often tagged with a polyhistidine tag (His tag) to facilitate purification. The production process involves the following steps:

  1. Gene Cloning: The gene encoding Complement Factor B is cloned into an expression vector.
  2. Transformation: The vector is introduced into the host cells (e.g., HEK293 or E. coli).
  3. Expression: The host cells are cultured under conditions that promote the expression of the recombinant protein.
  4. Purification: The recombinant protein is purified using affinity chromatography, exploiting the His tag for binding to a nickel or cobalt resin.
  5. Validation: The purity and activity of the recombinant protein are validated using techniques such as SDS-PAGE and functional assays.
Applications

Recombinant Complement Factor B is used in various research applications, including:

  • Studying the Complement System: Researchers use recombinant Complement Factor B to investigate the mechanisms of the alternative pathway and its role in immune responses.
  • Drug Development: It is used in the development of therapeutic agents targeting the complement system for diseases such as age-related macular degeneration and atypical hemolytic uremic syndrome.
  • Diagnostic Assays: Recombinant Complement Factor B is employed in diagnostic assays to measure complement activity and identify deficiencies or dysregulations in the complement pathway.

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.