FRZB Human

Frizzled-Related Protein Human Recombinant
Shipped with Ice Packs
In Stock

Description

Domain Architecture

FRZB contains two key domains:

  1. Cysteine-Rich Domain (CRD): Binds Wnt ligands, competing with Frizzled receptors to regulate canonical Wnt/β-catenin signaling .

  2. Netrin-like (NTN) Motif: Interacts with extracellular matrix components and influences non-canonical Wnt pathways (e.g., Wnt/Ca²⁺ signaling) .

DomainFunctionKey Interaction Partners
CRDWnt ligand bindingWnt proteins (e.g., Wnt3a, Wnt5a)
NTNECM interactionCollagen, proteoglycans

Recombinant FRZB Protein

  • Source: Produced in E. coli (non-glycosylated) .

  • Molecular Weight: 35 kDa (includes a 23-amino acid His-tag) .

  • Sequence: MGSSHHHHHHSSGLVPRGSH... (full sequence in ).

Role in Bone Development and Osteogenesis

FRZB promotes osteogenic differentiation by shifting Wnt signaling from canonical to non-canonical pathways:

Mechanistic Insights

  • Overexpression:

    • Reduces β-catenin levels (inhibits canonical Wnt) .

    • Activates Ca²⁺/CamKII signaling (enhances non-canonical Wnt) .

  • Loss of Function:

    • Suppresses osteoblast markers (e.g., Osterix, Osteocalcin) .

Experimental ModelOutcomeSignificance
MC3T3-E1 cells (mouse osteoblasts)↑ Alizarin Red staining, ↑ ALP activityFRZB enhances bone mineralization .
Human periosteal cells↑ Calcium deposition with FRZB-conditioned mediumTranslational potential in bone repair .

Osteoarthritis (OA)

  • Genetic Link: SNPs in FRZB (e.g., rs288326) correlate with female-specific OA susceptibility .

  • Mechanism: Dysregulation of chondrocyte maturation and cartilage homeostasis .

Muscle Denervation and ALS

  • Biomarker: ↑ FRZB in muscle biopsies of ALS patients, associated with atrophic fibers .

  • Pathway: FRZB antagonizes Wnt signaling, leading to ↑ β-catenin at myofiber membranes .

Osteomyelitis

  • Dual Role:

    • Downregulation: Reduces S. aureus-induced apoptosis, promotes osteogenic differentiation .

    • Upregulation: Enhances inflammation and inhibits bone repair .

Key Studies

Study TypeFocusFindingsImplications
In vitro (MC3T3-E1)OsteogenesisFRZB shifts Wnt signaling to non-canonical pathways Therapeutic target for bone disorders
Human cohort (OA)Genetic associationFRZB SNPs linked to female OA Gender-specific OA risk markers
ALS muscle biopsiesBiomarker discoveryFRZB correlates with disease progression Potential therapeutic or diagnostic target
S. aureus osteomyelitisInfection modelSilencing FRZB reduces inflammation FRZB inhibitors for osteomyelitis

Therapeutic Potential

  • Bone Disorders: FRZB agonists may enhance osteogenesis in osteoporosis or fracture healing .

  • Osteoarthritis: Targeting FRZB variants to mitigate cartilage degradation .

  • Osteomyelitis: FRZB modulation to balance anti-inflammatory and pro-repair effects .

Product Specs

Introduction
Frizzled-Related Protein (FRZB) is a secreted protein that plays a role in regulating bone development. Genetic defects in the FRZB gene are linked to an increased susceptibility to osteoarthritis (OA), particularly in females. FRZB belongs to the soluble frizzled-related protein (sFRP) family, which modulate Wnt signaling pathways by directly interacting with Wnt proteins. sFRPs are essential for regulating cell growth and differentiation in specific cell types. Notably, SFRP3/FRZB is involved in the development of limb skeletons. FRZB also regulates the maturation of chondrocytes and the growth of long bones.
Description
Recombinant human FRZB protein, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 316 amino acids (residues 33-325). It has a molecular weight of 35 kDa. The protein includes a 23 amino acid His-tag fused at the N-terminus to facilitate purification, which is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear and colorless solution that has been sterilized by filtration.
Formulation
The FRZB protein solution is provided at a concentration of 1 mg/ml in a buffer consisting of 20 mM Tris-HCl (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the FRZB protein solution should be kept refrigerated at 4°C. For extended storage, it is recommended to freeze the solution at -20°C. To ensure optimal protein stability during long-term storage, adding a carrier protein such as 0.1% HSA or BSA is advisable. Repeated freezing and thawing of the protein solution should be avoided.
Purity
The purity of the FRZB protein is greater than 90%, as determined by SDS-PAGE analysis.
Synonyms
Secreted frizzled-related protein 3, sFRP-3, Frezzled, Fritz, Frizzled-related protein 1, FrzB-1, FRZB, FIZ, FRE, FRP, FRZB1, SFRP3, OS1, FZRB, hFIZ, FRP-3, SFRP3, SRFP3, FRZB-PEN.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSAACEPVR IPLCKSLPWN MTKMPNHLHH STQANAILAI EQFEGLLGTH CSPDLLFFLC AMYAPICTID FQHEPIKPCK SVCERARQGC EPILIKYRHS WPENLACEEL PVYDRGVCIS PEAIVTADGA DFPMDSSNGN CRGASSERCK CKPIRATQKT YFRNNYNYVI RAKVKEIKTK CHDVTAVVEV KEILKSSLVN IPRDTVNLYT SSGCLCPPLN VNEEYIIMGY EDEERSRLLL VEGSIAEKWK DRLGKKVKRW DMKLRHLGLS KSDSSNSDST QSQKSGRNSN PRQARN.

Q&A

What is FRZB and what is its role in human biology?

FRZB (frizzled-related protein, also known as secreted frizzled-related protein-3 or SFRP3) is a Wnt signaling modulator that contains a characteristic amino-terminal cysteine-rich domain (CRD) and a carboxy-terminal Netrin-like (NTN) motif. The protein functions primarily as a modulator of Wnt signaling pathways, which are crucial for development, tissue homeostasis, and disease processes. In human biology, FRZB plays significant roles in skeletal development, bone metabolism, and joint biology. Studies have shown that FRZB can antagonize canonical Wnt signaling while potentially enhancing non-canonical Wnt pathways, creating a complex regulatory network that influences cell differentiation and tissue homeostasis . To investigate FRZB function in experimental settings, researchers typically use gain-of-function and loss-of-function approaches in relevant cell types, followed by assessment of downstream signaling events and phenotypic outcomes.

How is FRZB gene expression regulated in human tissues?

FRZB gene expression exhibits tissue-specific and developmental stage-specific patterns in humans. The gene is particularly expressed during skeletal development and in adult bone and cartilage tissues. Regulation occurs through multiple mechanisms including transcriptional control via promoter elements, epigenetic modifications, and post-transcriptional regulation. To study FRZB expression patterns, researchers employ quantitative PCR, in situ hybridization, and immunohistochemistry techniques on human tissue samples. For mechanistic studies of regulation, chromatin immunoprecipitation (ChIP) assays can identify transcription factor binding sites, while reporter gene assays can validate functional regulatory elements. When designing such experiments, it's important to include appropriate controls and consider the context-dependency of FRZB expression, as its regulation may differ significantly between tissue types and developmental stages .

What structural domains characterize the FRZB protein and how do they contribute to its function?

The FRZB protein contains two primary structural domains: an amino-terminal cysteine-rich domain (CRD) and a carboxy-terminal Netrin-like (NTN) motif. The CRD shares homology with the extracellular domain of Frizzled receptors and has traditionally been considered the primary Wnt-binding region. The NTN domain, while less studied, also contributes significantly to FRZB function, particularly in bone biology. To experimentally determine domain functions, researchers typically generate domain-specific deletion mutants (ΔCRD or ΔNTN) and assess their effects on Wnt binding, signaling outcomes, and biological processes such as osteogenesis. Studies have shown that full-length FRZB and NTN-only constructs produce similar effects on osteogenesis, whereas CRD-only constructs mimic loss-of-function phenotypes, suggesting domain-specific roles in regulating different aspects of Wnt signaling . For structural analysis, techniques such as X-ray crystallography, NMR spectroscopy, or cryo-EM can provide insights into protein-protein interactions and conformational changes upon Wnt binding.

What are the established methods for detecting FRZB protein in human samples?

Detection of FRZB protein in human samples can be accomplished through several complementary techniques. Western blotting using validated anti-FRZB antibodies remains the gold standard for protein quantification in tissue or cell lysates. For localization studies, immunohistochemistry or immunofluorescence on tissue sections or cultured cells provides spatial information. ELISA-based methods can quantify FRZB in serum, synovial fluid, or other biological fluids. When implementing these methods, researchers should:

  • Validate antibody specificity using positive and negative controls

  • Include recombinant FRZB protein standards for quantification

  • Consider protein extraction methods that preserve FRZB stability

  • Account for potential cross-reactivity with other SFRP family members

Mass spectrometry-based proteomic approaches offer an antibody-independent alternative and can provide additional information about post-translational modifications or protein interactions. Notably, detection sensitivity varies across methods, with ELISA typically offering the highest sensitivity for dilute samples .

How does FRZB differentially affect canonical versus non-canonical Wnt signaling pathways in human cells?

FRZB exhibits a complex, context-dependent regulatory effect on Wnt signaling pathways. Research has demonstrated that FRZB predominantly inhibits canonical Wnt/β-catenin signaling while simultaneously enhancing certain non-canonical Wnt pathways. This differential regulation creates a signaling switch that can profoundly impact cell fate decisions and tissue homeostasis. To investigate these effects experimentally, researchers should:

  • Perform dual-luciferase reporter assays using TOPFlash/FOPFlash for canonical signaling

  • Assess non-canonical pathway activation through phosphorylation of JNK, CAMKII, or PKC

  • Conduct parallel analyses in multiple cell types to capture context-dependency

  • Use dose-response experiments to identify potential biphasic effects

Western blot analysis in FRZB-overexpressing or knockout cell lines shows reduced β-catenin nuclear translocation and target gene expression for canonical pathways, while simultaneously revealing increased phosphorylation of non-canonical pathway components. This dual regulatory role may explain the seemingly contradictory phenotypes observed in different physiological contexts . When interpreting results, researchers should consider that the ratio of canonical to non-canonical signaling may be more important than absolute levels of each pathway's activation.

What experimental models are most appropriate for studying FRZB function in human bone and joint disorders?

The selection of experimental models for studying FRZB in bone and joint disorders requires careful consideration of translational relevance and methodological limitations. The following hierarchy of models provides increasing physiological relevance:

Model TypeAdvantagesLimitationsKey Applications
Cell lines (MC3T3-E1, hBMSCs)Controlled environment, genetic manipulationLack tissue complexityMolecular mechanisms, signaling
Primary human cellsHuman relevance, donor-specific responsesLimited availability, variabilityTranslational validation
Ex vivo tissue culturesMaintains tissue architectureShort-term viabilityEarly drug screening
Frzb−/− miceSystemic effects, developmental insightsSpecies differencesPhenotypic analysis, pathway discovery
Human tissue samplesDirect clinical relevanceLimited experimental manipulationBiomarker validation, expression studies

Research has demonstrated that conditioned medium from FRZB-overexpressing cells enhances osteogenesis in human periosteal and bone marrow stromal cells, validating translational relevance between models . When designing studies, researchers should consider using multiple complementary models and prioritize validation in primary human cells whenever possible. For joint disorders specifically, explant cultures of human cartilage combined with mechanical loading systems can provide valuable insights into FRZB's role in maintaining tissue homeostasis under physiological stress.

How can researchers distinguish between direct and indirect effects of FRZB on osteogenesis in experimental settings?

Distinguishing direct from indirect effects of FRZB on osteogenesis presents a significant methodological challenge. To address this, researchers should implement a systematic experimental approach:

  • Temporal analysis: Map the sequence of molecular events following FRZB manipulation using time-course experiments to establish causality

  • Molecular inhibition studies: Selectively block potential mediators to identify essential signaling nodes

  • Proximity-based interaction studies: Use techniques like proximity ligation assay (PLA) or FRET to detect direct FRZB-protein interactions

  • Domain-specific constructs: Compare effects of full-length FRZB versus CRD-only or NTN-only variants

  • Transcriptomics with network analysis: Identify immediate-early response genes versus secondary transcriptional changes

What are the methodological considerations for studying FRZB polymorphisms in relation to human bone mass variation?

Investigating associations between FRZB polymorphisms and bone mass phenotypes requires rigorous methodological approaches to ensure valid, reproducible findings. Researchers should consider the following framework:

  • Study design considerations:

    • Power calculations based on expected effect sizes (typically modest for complex traits)

    • Population stratification control through genomic methods

    • Replication in independent cohorts with similar demographic profiles

    • Longitudinal approaches to capture age-related changes in bone parameters

  • Genotyping approaches:

    • Targeted SNP analysis focusing on functional variants (coding regions, splice sites)

    • Haplotype analysis rather than isolated SNPs

    • Next-generation sequencing for rare variant detection

    • Evaluation of copy number variations

  • Phenotyping precision:

    • Standardized DXA measurements at multiple skeletal sites

    • Advanced imaging (pQCT, HR-pQCT) for volumetric and microstructural analysis

    • Biochemical markers of bone turnover as intermediate phenotypes

    • Detailed medical history to account for confounding factors

  • Functional validation of identified variants:

    • In vitro mutagenesis to assess effects on protein structure and function

    • Cell-based assays to determine impact on Wnt signaling efficiency

    • CRISPR-edited cellular models expressing variant forms

When interpreting results, researchers should consider that FRZB polymorphisms likely interact with other genetic factors and environmental influences to determine bone mass. The complex nature of bone homeostasis means that isolated polymorphisms typically explain only a small portion of phenotypic variance, necessitating large sample sizes and careful statistical approaches .

How can researchers effectively evaluate the therapeutic potential of FRZB modulation in human bone disorders?

Evaluating FRZB's therapeutic potential for bone disorders requires a comprehensive translational research pipeline that bridges basic science discoveries with clinical applications. A methodological framework should include:

  • Target validation phase:

    • Confirmation of FRZB's role in disease-relevant human tissues

    • Identification of disease-specific expression patterns or modifications

    • Determination of upstream regulators and downstream effectors

  • Intervention strategy development:

    • Recombinant protein administration versus gene therapy approaches

    • Domain-specific therapies (CRD versus NTN) for pathway-selective effects

    • Local versus systemic delivery methods to minimize off-target effects

  • Preclinical efficacy assessment:

    • Dose-finding studies in relevant cell culture systems

    • Ex vivo human bone explant cultures for rapid translational insights

    • Animal models with humanized bone components when possible

  • Safety and pharmacology evaluation:

    • Assessment of effects on multiple Wnt-dependent tissues

    • Long-term effects on bone remodeling and quality

    • Potential compensatory mechanisms from other SFRP family members

Research indicates that FRZB and other SFRPs or their specific domains may have unexpected therapeutic potential for bone and joint disorders, as excess SFRPs can produce effects not observed under physiological conditions . When developing therapeutic approaches, researchers should carefully consider the paradoxical effects observed in different contexts—FRZB can both enhance osteogenesis through non-canonical Wnt pathways while inhibiting canonical Wnt signaling. This dual action may allow for context-specific therapeutic applications but requires precise understanding of the molecular mechanisms in each disorder.

What are the optimal protocols for isolating and purifying recombinant FRZB protein for functional studies?

The production of high-quality recombinant FRZB protein is critical for functional studies but presents several technical challenges due to its complex domain structure and potential for misfolding. A comprehensive protocol should address:

  • Expression system selection:

    • Mammalian expression systems (HEK293, CHO cells) provide superior folding and post-translational modifications compared to bacterial systems

    • Insect cell systems (Sf9, High Five) offer a balance of proper folding and higher yield

    • Avoid bacterial expression systems that typically yield improperly folded protein with compromised activity

  • Vector design considerations:

    • Inclusion of a cleavable secretion signal for extracellular accumulation

    • Addition of purification tags (His6, FLAG) positioned to avoid interference with functional domains

    • Codon optimization for the selected expression system

    • Optional inclusion of domain-specific constructs (CRD-only, NTN-only) for comparative studies

  • Purification strategy:

    • Two-step chromatography combining affinity purification and size exclusion

    • Careful buffer optimization to maintain protein stability (typically pH 7.4-8.0 with low salt)

    • Inclusion of protease inhibitors throughout purification process

    • Quality control via SDS-PAGE, Western blot, and activity assays

  • Storage and stability:

    • Flash freezing in small aliquots with cryoprotectants (5-10% glycerol)

    • Stability testing at different temperatures and after freeze-thaw cycles

    • Activity assays before experimental use to ensure functional integrity

When conducting functional studies with purified FRZB, researchers should include proper controls such as heat-denatured protein and unrelated proteins of similar size to distinguish specific from non-specific effects. Functional validation should include Wnt-binding assays to confirm that the recombinant protein retains its native binding properties .

How can researchers effectively differentiate between the roles of FRZB and other SFRP family members in experimental settings?

Distinguishing the specific functions of FRZB from other SFRP family members requires targeted experimental approaches that account for their structural similarities and potential functional redundancy. A methodological framework should include:

  • Expression analysis specificity:

    • Use of validated primers/probes with demonstrated lack of cross-reactivity

    • Correlation of mRNA and protein levels to account for post-transcriptional regulation

    • Single-cell RNA sequencing to identify cell populations expressing specific SFRPs

  • Loss-of-function strategies:

    • CRISPR-Cas9 gene editing for complete knockout studies

    • siRNA or shRNA with demonstrated specificity and efficiency

    • Combination knockouts to identify redundant functions

    • Domain-specific deletions to identify unique functional regions

  • Gain-of-function approaches:

    • Controlled expression systems with physiologically relevant levels

    • Domain swapping between SFRPs to identify functional determinants

    • Conditioned media approaches with quantification of secreted protein

  • Interaction profiling:

    • Comparative Wnt binding assays to identify differential affinities

    • Co-immunoprecipitation studies with controls for non-specific binding

    • Surface plasmon resonance to determine binding kinetics and affinities

Research has demonstrated that different SFRP family members can have distinct or even opposing effects on Wnt signaling and downstream biological processes. For example, while FRZB may promote osteogenesis in certain contexts, other SFRPs might inhibit it . When designing experiments, researchers should consider the expression profiles of all SFRP family members in their experimental system and account for potential compensatory mechanisms that may occur when manipulating individual SFRPs.

What technical considerations are essential when studying FRZB-mediated effects on Wnt signaling pathways?

Investigating FRZB's effects on Wnt signaling requires sophisticated technical approaches that account for the complexity and context-dependency of these pathways. Key methodological considerations include:

  • Pathway activation controls:

    • Use of appropriate Wnt ligands at physiological concentrations

    • Inclusion of positive controls (direct pathway activators) and negative controls

    • Time-course experiments to capture transient signaling events

    • Assessment of multiple pathway components rather than single nodes

  • Reporter system selection:

    • TOPFlash/FOPFlash for canonical pathway activity

    • Non-canonical pathway reporters (NFAT, AP-1) when appropriate

    • Internal normalization controls to account for cell number and transfection efficiency

    • Consideration of endogenous pathway activity in the chosen cell type

  • Protein-level analysis:

    • Fractionation techniques to assess β-catenin nuclear translocation

    • Phosphorylation-specific antibodies for non-canonical pathway components

    • Co-immunoprecipitation to detect physical interactions between FRZB and Wnt ligands

    • Proximity ligation assays to visualize protein interactions in situ

  • Transcriptional target assessment:

    • Selection of well-validated Wnt target genes for qPCR analysis

    • Consideration of cell-type specificity in target gene selection

    • RNA-seq to identify global transcriptional changes

    • ChIP-seq for β-catenin or TCF/LEF binding to identify direct targets

Research has shown that FRZB overexpression results in reduced canonical but increased non-canonical Wnt signaling, while FRZB loss produces the opposite effects . This dual regulatory role complicates interpretation and requires simultaneous assessment of multiple pathway branches. When designing experiments, researchers should consider that the effects of FRZB may vary significantly depending on the complement of Wnt ligands, receptors, and co-receptors expressed in the experimental system, necessitating careful characterization of baseline pathway components.

What are the most reliable biomarkers for monitoring FRZB activity in human clinical samples?

Establishing reliable biomarkers for FRZB activity in clinical samples is essential for translational research and potential therapeutic development. A comprehensive biomarker strategy should consider:

The clinical utility of these biomarkers depends on their sensitivity, specificity, and correlation with disease processes. In translational studies, researchers have observed that FRZB levels in synovial fluid correlate with joint pathology, while serum levels may reflect systemic bone metabolism . When developing biomarker panels, researchers should consider combining direct FRZB measurements with downstream pathway indicators to provide a more complete picture of FRZB activity in the tissue or system of interest.

What emerging technologies could advance our understanding of FRZB function in human biology?

Several cutting-edge technologies hold promise for deeper insights into FRZB biology and function:

  • Spatial transcriptomics and proteomics:

    • Single-cell spatial analysis to map FRZB expression and activity in tissue microenvironments

    • Multiplexed protein imaging to visualize FRZB interactions with Wnt pathway components in situ

    • Integration of transcriptomic and proteomic data for comprehensive pathway mapping

  • Advanced genetic engineering:

    • CRISPR base editing for introducing precise polymorphisms without double-strand breaks

    • Conditional and tissue-specific gene manipulation in human organoids

    • Synthetic biology approaches to create tunable FRZB expression systems

  • Structural biology innovations:

    • Cryo-EM analysis of FRZB-Wnt complexes at near-atomic resolution

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic protein interactions

    • Computational modeling and molecular dynamics simulations of domain-specific functions

  • Translational research platforms:

    • Patient-derived iPSC differentiation to bone and cartilage lineages

    • Organ-on-chip technologies to model FRZB function in bone-cartilage interfaces

    • Humanized mouse models with patient-specific FRZB variants

These technologies could help address key knowledge gaps, such as the atomic-level interactions between FRZB domains and specific Wnt ligands, the tissue-specific regulation of FRZB expression, and the integration of FRZB signaling with other pathways in complex bone and joint disorders . Researchers should consider forming interdisciplinary collaborations to leverage these technologies effectively, as they often require specialized expertise across molecular biology, bioengineering, computational science, and clinical research domains.

How should researchers approach contradictory findings in the FRZB literature?

The FRZB research field contains several apparent contradictions that require careful methodological approaches to resolve. A systematic framework for addressing these inconsistencies includes:

  • Context evaluation:

    • Detailed comparison of experimental systems (cell types, species, developmental stages)

    • Assessment of baseline Wnt pathway component expression

    • Consideration of dose-dependent effects that may produce biphasic responses

    • Evaluation of acute versus chronic FRZB manipulation

  • Methodological standardization:

    • Development of common assay protocols and reagents

    • Reporting of detailed experimental conditions and controls

    • Use of multiple complementary techniques to measure the same outcome

    • Independent replication in different laboratories

  • Data integration approaches:

    • Meta-analysis of published findings with attention to methodological differences

    • Network analysis to identify context-specific signaling nodes

    • Development of computational models that can predict context-dependent outcomes

    • Integration of in vitro, animal model, and human data

  • Targeted hypothesis testing:

    • Design of experiments specifically aimed at resolving contradictions

    • Use of domain-specific constructs to identify region-dependent effects

    • Direct comparison of competing models in identical experimental systems

Research has shown that FRZB can both promote and inhibit osteogenesis depending on context, likely through differential regulation of canonical versus non-canonical Wnt pathways . When approaching contradictory findings, researchers should consider that these may reflect true biological complexity rather than experimental artifacts, and design studies that can explore the mechanisms underlying context-dependent effects. Collaborative approaches, including pre-registered multi-laboratory studies, can be particularly valuable for resolving persistent contradictions in the literature.

What are the key considerations for translating basic FRZB research findings to clinical applications?

Translating fundamental discoveries about FRZB biology into clinical applications requires addressing several critical challenges:

  • Target validation in human contexts:

    • Confirmation of findings in primary human cells and tissues

    • Correlation of FRZB expression/activity with clinical outcomes in patient cohorts

    • Identification of patient subgroups most likely to benefit from FRZB-targeted therapies

    • Assessment of potential compensatory mechanisms in chronic conditions

  • Therapeutic development considerations:

    • Domain-specific approaches to selectively modulate specific Wnt pathways

    • Tissue-targeted delivery systems to minimize systemic effects

    • Stability and pharmacokinetic optimization of FRZB-based biologics

    • Development of small molecule modulators of FRZB activity

  • Preclinical to clinical transition:

    • Selection of relevant animal models that recapitulate human pathophysiology

    • Identification and validation of translational biomarkers

    • Early engagement with regulatory agencies regarding novel modalities

    • Consideration of combination therapies with existing standard of care

  • Clinical trial design elements:

    • Patient stratification based on FRZB polymorphisms or baseline expression

    • Selection of relevant clinical endpoints and biomarkers

    • Determination of optimal timing for intervention in disease course

    • Long-term safety monitoring for effects on multiple Wnt-dependent tissues

Research suggests that FRZB and other SFRPs may have unexpected therapeutic potential in bone and joint disorders because excess levels can produce effects not observed under physiological conditions . This opens opportunities for therapeutic manipulation but requires precise understanding of dose-response relationships and contextual factors. When developing translational programs, researchers should consider that the complex dual role of FRZB in regulating different Wnt pathways may allow for highly specific interventions but also creates challenges for predicting outcomes across different tissues and disease states.

Product Science Overview

Structure and Function

Frizzled-related proteins contain a cysteine-rich domain (CRD) that is homologous to the Wnt-binding domain of Frizzled receptors . This domain enables FRPs to bind to Wnt ligands or Frizzled receptors, thereby modulating Wnt signaling . The human recombinant version of these proteins is produced using recombinant DNA technology, which involves inserting the gene encoding the protein into a suitable expression system, such as bacteria or mammalian cells, to produce the protein in large quantities .

Role in Wnt Signaling

The Wnt signaling pathway can be divided into two main branches: the canonical (β-catenin-dependent) pathway and the non-canonical (β-catenin-independent) pathway . FRPs can act as antagonists of the Wnt signaling pathway by binding to Wnt ligands and preventing them from interacting with Frizzled receptors . This inhibition can affect various cellular processes, including cell proliferation, differentiation, and migration .

Applications in Research and Medicine

Recombinant human FRPs are valuable tools in research and medicine due to their ability to modulate Wnt signaling . They are used in various applications, including:

  • Cancer Research: FRPs have been shown to play a role in tumor angiogenesis, making them potential targets for anti-cancer therapies .
  • Developmental Biology: Studying the role of FRPs in embryonic development can provide insights into the mechanisms underlying various developmental disorders .
  • Regenerative Medicine: Modulating Wnt signaling using FRPs can promote tissue regeneration and repair .

In conclusion, Frizzled-related proteins are essential regulators of the Wnt signaling pathway, with significant implications for developmental biology, cancer research, and regenerative medicine. The recombinant production of these proteins has enabled their widespread use in various research and therapeutic applications.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.