OPG Human, Hi-5

Osteoprotegerin Human Recombinant, Hi-5
Shipped with Ice Packs
In Stock

Description

Role of OPG in Bone Metabolism

OPG is pivotal in maintaining bone homeostasis by regulating osteoclast activity. Osteoclasts are responsible for bone resorption, a process that is balanced by bone formation. By binding to RANKL, OPG prevents the interaction between RANKL and its receptor RANK, thus inhibiting osteoclast differentiation and activation . This mechanism is crucial for preventing excessive bone loss, which can lead to conditions like osteoporosis.

OPG in Disease Pathogenesis

Beyond its role in bone health, OPG is implicated in various diseases:

  • Atherosclerosis and Aneurysms: OPG is associated with the growth of abdominal aortic aneurysms (AAA) and atherosclerosis. It influences vascular smooth muscle cell proliferation and apoptosis, contributing to aneurysm progression .

  • Cancer: OPG can inhibit apoptosis in tumor cells by binding to TRAIL, potentially promoting tumor survival and metastasis .

  • Immune Regulation: OPG plays a role in immune system development and signaling, affecting inflammatory processes .

Research Findings and Data

While specific data on "OPG Human, Hi-5" is limited, research on human osteoprotegerin provides valuable insights into its function and measurement:

ELISA Kits for OPG Measurement

ELISA kits are commonly used to quantify OPG levels in serum and plasma samples. These kits typically employ a sandwich ELISA technique, where OPG is captured by immobilized antibodies and then detected using biotin-labeled antibodies .

Sample TypeAverage % RecoveryRange
Serum9488-99
EDTA Plasma9587-100
Citrate Plasma8882-96
Heparin Plasma9167-118

Table 1: Recovery of Recombinant OPG in Different Matrices

OPG in Disease Models

Studies using OPG-Fc fusion proteins have shown that high doses can lead to osteopetrosis-like changes in mice, characterized by reduced osteoclast activity and bone modeling defects .

TreatmentTRACP-5b Activity (U/L)
Saline1.99 ± 0.42
Low Dose OPG-Fc0.71 ± 0.56
High Dose OPG-Fc0.50 ± 0.35

Table 2: Effect of OPG-Fc Treatment on TRACP-5b Activity

Product Specs

Introduction
Osteoprotegerin (OPG) is a soluble protein that plays a crucial role in bone remodeling by acting as a decoy receptor for RANKL. By binding to RANKL, OPG prevents the interaction between RANKL and its receptor RANK, thereby inhibiting the differentiation and activation of osteoclasts, the cells responsible for bone resorption. This inhibitory effect on osteoclastogenesis helps maintain bone density and prevent bone loss. OPG also appears to have a protective role in vascular calcification and may safeguard cells from apoptosis induced by the protein TRAIL.
Description
Recombinant Osteoprotegerin, expressed in baculovirus, is a single, glycosylated polypeptide chain consisting of 398 amino acid residues (22-401 a.a.). It has a calculated molecular mass of 45.6 kDa. This OPG protein is engineered with a 17 amino acid His tag at the C-terminus to facilitate purification, which is achieved using proprietary chromatographic techniques.
Physical Appearance
The product is a sterile, filtered solution that is colorless and clear.
Formulation
Osteoprotegerin protein is supplied in PBS buffer with a pH of 7.4 and 10% glycerol.
Stability
For short-term storage (2-4 weeks), the product should be kept at 4°C. For long-term storage, it is recommended to store the product frozen at -20°C. Avoid repeated freeze-thaw cycles.
Purity
The purity of the Osteoprotegerin protein is greater than 85.0% as determined by SDS-PAGE analysis.
Synonyms
TNFRSF11B, OPG, OCIF, Osteoclastogenesis inhibitory factor, Osteoprotegerin, TR1, MGC29565.
Source
Hi-5.
Amino Acid Sequence
ADPETFPPKY LHYDEETSHQ LLCDKCPPGT YLKQHCTAKW KTVCAPCPDH YYTDSWHTSD ECLYCSPVCK ELQYVKQECN RTHNRVCECK EGRYLEIEFC LKHRSCPPGF GVVQAGTPER NTVCKRCPDG FFSNETSSKA PCRKHTNCSV FGLLLTQKGN ATHDNICSGN SESTQKCGID VTLCEEAFFR FAVPTKFTPNWLSVLVDNLP GTKVNAESVE RIKRQHSSQE QTFQLLKLWK HQNKDQDIVK KIIQDIDLCE NSVQRHIGHA NLTFEQLRSL MESLPGKKVG AEDIEKTIKACKPSDQILKL LSLWRIKNGD QDTLKGLMHA LKHSKTYHFP KTVTQSLKKT IRFLHSFTMY KLYQKLFLEM IGNQVQSVKI SCLSGRLVPR GSHHHHHH.

Q&A

What is Osteoprotegerin (OPG) and what functional roles does it play in human physiology?

Osteoprotegerin (OPG), also known as TNFRSF11B, is a secreted glycoprotein member of the tumor necrosis factor receptor superfamily. OPG functions primarily as a decoy receptor for RANKL (Receptor Activator of Nuclear Factor κB Ligand), thereby neutralizing its function in osteoclastogenesis. This interaction inhibits osteoclast activation and promotes osteoclast apoptosis, making OPG a critical regulator of bone homeostasis .

Beyond bone metabolism, OPG exhibits diverse physiological functions:

  • Acts as a decoy receptor for TRAIL (TNF-related apoptosis-inducing ligand), potentially protecting cells from apoptosis

  • Plays roles in immune response regulation via RANKL interactions with dendritic cells

  • Contributes to vascular health, with associations to pathologies like coronary artery disease, atherosclerosis, and vascular calcification

  • Demonstrates involvement in chondrocyte metabolism and osteoarthritis progression

The OPG/RANKL ratio is considered a critical determinant in bone remodeling, with altered ratios observed in pathological conditions such as osteoarthritis .

What are High Five (Hi-5) cells and why are they advantageous for recombinant protein expression?

High Five cells (BTI-TN-5B1-4) are an insect cell line derived from the cabbage looper Trichoplusia ni ovary, developed by the Boyce Thompson Institute for Plant Research . These cells have become a preferred expression system for recombinant proteins due to several key advantages:

  • Superior secretion capacity: High Five cells provide up to 10-fold higher secreted protein expression compared to Sf9 cells

  • Growth flexibility: Can be cultured in both adherent and suspension formats

  • Serum-free adaptation: Successfully grow in serum-free media, reducing purification complexity

  • Exceptional protein production rate: Exhibit specific protein production rates of 5.1 × 10⁻⁶ µg/(cell·h), the highest among nine insect cell lines tested in comparative studies

  • Compatibility: Work effectively with multiple baculovirus expression systems including BaculoDirect™, Bac-to-Bac™, and InsectDirect™

These characteristics make High Five cells particularly valuable for researchers seeking high-yield production of complex secreted proteins like human OPG.

How does the OPG/RANK/RANKL axis function in bone metabolism and disease?

The OPG/RANK/RANKL triad constitutes a molecular regulatory system central to bone homeostasis and remodeling:

Functional relationships:

  • RANKL (expressed by osteoblasts and activated T cells) binds to RANK on osteoclast precursors, stimulating osteoclastogenesis and bone resorption

  • OPG acts as a soluble decoy receptor for RANKL, preventing RANKL-RANK binding and inhibiting osteoclast formation and function

  • The OPG/RANKL ratio critically determines bone resorption rates

Pathological alterations:

  • In osteoarthritis, the OPG/RANKL ratio is significantly reduced on chondrocytes (p=0.05), while the RANK/RANKL ratio is significantly increased (p<0.03)

  • Inflammatory cytokines (IL-1β, TNF-α, PGE₂) significantly enhance OPG and membranous RANKL levels in chondrocytes, whereas only IL-1β significantly increases membranous RANK

  • Paradoxically, OPG can promote pathological progression in osteoarthritis by stimulating MMP-13 (p=0.05) and protease-activated receptor-2 (PAR-2) (p<0.04) production

The relative ratio of RANKL/OPG has been demonstrated as significant to processes such as odontoclastogenesis, with OPG protecting dental pulp from damage-induced odontoclast formation .

What are the optimal expression conditions for producing recombinant human OPG in Hi-5 cells?

For optimal expression of recombinant human OPG in High Five cells, researchers should consider the following methodological approach:

Expression system selection:

  • Baculovirus expression system using Hi-5 cells consistently yields higher secreted OPG than alternative systems

  • Using a baculovirus vector containing human OPG cDNA (amino acids 22-401, corresponding to the mature protein without the signal peptide)

Expression construct design:

  • Include a C-terminal His-tag for purification purposes (typically 6-8 histidine residues)

  • The complete amino acid sequence should include positions 22-401 (ETFPPKYLHYDEETSHQ...CKRCPDGFFSNETSSKAPCRKHTNCSV)

Culture conditions:

  • Maintain cells in serum-free medium such as Express Five™ SFM

  • For suspension culture: 27-28°C with shaking at 120-150 rpm

  • Cell density should be maintained between 0.5-6.0 × 10⁶ cells/mL for optimal protein expression

  • Harvest 72-96 hours post-infection when protein production peaks

Expected yields:

  • High Five cells can produce OPG at concentrations of 20-50 mg/L of culture

  • Specific production rate of approximately 5.1 × 10⁻⁶ µg/(cell·h)

This methodology capitalizes on Hi-5 cells' exceptional secretory capacity while providing a fusion-tagged product that facilitates downstream purification.

What purification strategies are most effective for isolating human OPG from Hi-5 cell cultures?

Purification of His-tagged human OPG from Hi-5 cell culture supernatant typically follows a multi-step chromatographic approach:

Primary capture step:

  • Immobilized metal affinity chromatography (IMAC) using Ni-NTA resin to capture the His-tagged OPG

  • Load filtered culture supernatant directly onto equilibrated column

  • Wash with PBS containing 10-20 mM imidazole to reduce non-specific binding

  • Elute with imidazole gradient (50-250 mM) in PBS

Secondary purification:

  • Size exclusion chromatography to remove aggregates and achieve higher purity

  • Ion exchange chromatography (typically anion exchange) to remove remaining contaminants

Final formulation:

  • Buffer exchange into storage buffer: PBS (pH 7.4) containing 10% glycerol

  • Filter sterilization (0.22 μm)

  • Quality control typically reveals >90% purity by SDS-PAGE and endotoxin levels <1 EU per 1μg of protein

Critical considerations:

  • OPG is a glycoprotein with apparent molecular weight of ~55 kDa on SDS-PAGE despite a calculated mass of ~44.7 kDa (389aa)

  • Inclusion of protease inhibitors during initial processing steps is recommended

  • For long-term storage, aliquoting and storage at -20°C to -80°C with avoidance of freeze-thaw cycles is advised

This purification strategy typically yields recombinant human OPG with appropriate structural and functional characteristics for research applications.

What analytical methods are recommended for characterizing recombinant human OPG from Hi-5 cells?

Comprehensive characterization of Hi-5-produced human OPG requires multiple analytical approaches to assess identity, purity, structure, and function:

Physicochemical characterization:

  • SDS-PAGE: Confirms molecular weight (~55 kDa for glycosylated form) and purity (target >90%)

  • Western blot: Verifies identity using anti-OPG or anti-His antibodies

  • Mass spectrometry: Provides accurate mass and can confirm amino acid sequence through peptide mapping

  • Isoelectric focusing: Determines charge heterogeneity and isoelectric point

Structural analysis:

  • Circular dichroism: Evaluates secondary structure elements

  • Size exclusion chromatography: Assesses aggregation state and homogeneity

  • Glycan analysis: Characterizes N-linked glycosylation patterns unique to insect cell expression

Functional testing:

  • Binding assays: Determines affinity for RANKL and TRAIL using surface plasmon resonance

  • Bioactivity determination: Measures ability to inhibit cytotoxicity using Jurkat human acute T cell leukemia cells in the presence of human TRAIL (typical ED50 ≤ 8 ng/ml)

  • Osteoclastogenesis inhibition assay: Confirms biological activity in primary osteoclast precursor cultures

Contaminant analysis:

  • Host cell protein ELISA: Quantifies residual insect cell proteins

  • Endotoxin testing (LAL method): Should be <1 EU per 1μg of protein

  • DNA quantification: Measures residual DNA from expression system

This multi-faceted analytical approach ensures that the recombinant OPG meets the rigorous standards required for research applications.

How is recombinant human OPG used to study the pathophysiology of osteoarthritis?

Recombinant human OPG serves as a valuable tool for investigating osteoarthritis (OA) pathophysiology through several experimental approaches:

Molecular interaction studies:

  • Examining how OPG interacts with other molecules involved in cartilage degradation

  • Research has revealed that OPG administration to OA chondrocytes significantly stimulates production of catabolic factors including MMP-13 (p=0.05) and protease-activated receptor-2 (PAR-2) (p<0.04)

Cytokine modulation experiments:

  • Investigating how inflammatory mediators affect OPG/RANKL/RANK expression

  • Studies show that IL-1β, TNF-α, and PGE₂ significantly enhance OPG and membranous RANKL levels in chondrocytes, while only IL-1β significantly increases membranous RANK expression

OPG/RANKL ratio analysis:

  • Measuring altered ratios in disease states compared to healthy controls

  • OA chondrocytes demonstrate significantly reduced OPG/RANKL ratio (p=0.05) and significantly increased RANK/RANKL ratio (p<0.03) compared to normal chondrocytes

Expression pattern analysis:

  • Assessing differential expression in affected tissues

  • Normal and OA chondrocytes express all three factors (OPG, RANK, RANKL), with OPG expression levels approximately 10³ and 10⁵ times higher than RANKL and RANK, respectively

These research applications of recombinant OPG have enhanced our understanding of OA as not merely a wear-and-tear condition but a complex metabolic disorder involving altered bone and cartilage homeostasis.

What role do OPG polymorphisms play in bone disorders and how can researchers study them?

OPG genetic polymorphisms significantly impact bone metabolism and disease susceptibility, offering researchers important targets for investigation:

Significant polymorphisms:

  • rs2073618 (G1181C): The CC genotype carriers show 2.18 times higher risk of peri-implantitis than GG genotype carriers (OR=2.18, 95% CI=1.03–4.62, p=0.04)

  • rs2073617 (T950C): Shows less consistent association with bone disorders

  • Haplotype analysis reveals that the G-C haplotype of rs2073618-rs2073617 correlates significantly with increased peri-implantitis susceptibility (OR=2.27, 95% CI=1.20–4.30)

Methodological approaches:

  • PCR-RFLP (polymerase chain reaction-restriction fragment length polymorphism) for genotyping OPG polymorphisms

  • Primer sequences for amplification:

    SNPPrimer sequenceTm (°C)
    rs2073618Forward: 5′-CCAAGCCCCTGAGGTTT-3′68°C
    Reverse: 5′-GGAGACCAGGTGGCAGC-3′
    rs2073617Forward: 5′-CCTGGGGGATCCTTTCC-3′54°C
    Reverse: 5′-AAGTATCGCCTGCCTTTGA-3′
  • Linkage disequilibrium (LD) and haplotype analysis performed with Haploview software

Research applications:

  • Case-control studies to determine polymorphism associations with specific bone disorders

  • Functional studies using site-directed mutagenesis of recombinant OPG to mimic polymorphic variants

  • Comparing polymorphism frequencies across different populations using Hardy-Weinberg equilibrium testing

  • Correlating genotypes with clinical parameters of bone metabolism and disease progression

These approaches enable researchers to elucidate how genetic variations in OPG contribute to altered bone homeostasis and disease susceptibility, potentially leading to personalized interventions.

How does the OPG/RANKL ratio function as a biomarker in bone and inflammatory disorders?

The OPG/RANKL ratio serves as a crucial biomarker reflecting bone remodeling balance, with significant implications for various disorders:

Quantification methods:

  • ELISA-based measurement of OPG in human serum/plasma:

    • Sensitivity: 0.07 pmol/l (≈1.4 pg/ml)

    • Standard range: 0–20 pmol/l (≈0–400 pg/ml)

    • Conversion factor: 1 pg/ml = 0.05 pmol/l (MW: 19.9 kDa)

  • qPCR for mRNA expression analysis using primers specific to human OPG and RANKL genes

Diagnostic significance:

  • Decreased OPG/RANKL ratio in OA chondrocytes (p=0.05) compared to normal

  • Elevated RANK/RANKL ratio in OA (p<0.03)

  • Decreased OPG/RANKL ratio in dental pulp tissue following traumatic injury

  • Studies reveal that human OPG expression levels are approximately 10³ and 10⁵ times higher than RANKL and RANK expression, respectively

Clinical correlations:

  • In falciparum malaria, OPG concentrations correlate with parasitemia, age, creatinine, lactate, and endothelial activation markers (angiopoietin-2, ICAM-1, E-selectin)

  • OPG is increased in patients with falciparum and vivax malaria compared to controls (p<0.0001), with higher levels in those with severe disease

  • The ligands RANKL and TRAIL are reduced in adults with malaria, with TRAIL reduction associated with disease severity markers

Mechanistic insights:

  • Inflammatory cytokines (IL-1β, TNF-α, PGE₂) significantly enhance OPG levels, potentially as a compensatory mechanism

  • Hypoxia-inducible factors (HIF-1α and HIF-2α) regulate RANKL/OPG expression, with HIF-1α reported to increase RANKL and decrease OPG

The OPG/RANKL ratio thus provides researchers with a quantifiable measure of bone metabolism disturbance applicable across multiple disease states.

What are the critical differences between recombinant OPG produced in Hi-5 cells versus mammalian systems?

Recombinant human OPG produced in Hi-5 cells differs from mammalian-expressed OPG in several important aspects that researchers must consider:

Glycosylation patterns:

  • Hi-5 cells produce simpler, high-mannose N-linked glycans lacking terminal sialic acids present in mammalian cells

  • Insect-derived glycosylation may alter protein half-life in circulation and potentially affect immunogenicity

  • Apparent molecular weight of glycosylated rhOPG protein from Hi-5 cells is approximately 55 kDa on SDS-PAGE despite a calculated mass of 44.7 kDa (389aa)

Protein yield advantages:

  • Hi-5 cells provide 5-10 fold higher secreted protein expression compared to mammalian cells and even other insect cells (Sf9)

  • High Five cells exhibit specific protein production rates of 5.1 × 10⁻⁶ µg/(cell·h), the highest among nine insect cell lines tested

Structural considerations:

  • Despite glycosylation differences, the core protein maintains proper folding for bioactivity

  • Biological activity assays show Hi-5-produced OPG effectively inhibits cytotoxicity in Jurkat cells (ED50 ≤ 8 ng/ml)

Production parameters:

  • Hi-5 expression system offers more rapid production cycles (72-96 hours post-infection)

  • Lower production costs compared to mammalian systems

  • Serum-free adaptation simplifies downstream purification

Research applications:

  • Most suitable for in vitro studies and fundamental research

  • Considerations for immunological studies where glycosylation differences may be significant

  • Appropriate for structural studies and ligand-binding experiments

Understanding these differences allows researchers to select the optimal expression system based on their specific experimental requirements and downstream applications.

How do inflammatory cytokines modulate the OPG/RANK/RANKL system in different tissue types?

Inflammatory cytokines exert tissue-specific modulatory effects on the OPG/RANK/RANKL system, creating complex patterns of regulation:

Chondrocyte responses:

  • IL-1β, TNF-α, and PGE₂ significantly enhance both OPG and membranous RANKL levels

  • IL-1β uniquely increases membranous RANK expression

  • These changes may contribute to cartilage degradation, as OPG administration to OA chondrocytes stimulates catabolic factors MMP-13 (p=0.05) and PAR-2 (p<0.04)

Osteoblast responses:

  • In human OA subchondral bone osteoblasts:

    • Two subpopulations exist based on PGE₂ production (low [L] or high [H])

    • L OA osteoblasts express significantly less OPG (p<0.03) than H OA osteoblasts

    • L OA osteoblasts show higher RANKL expression than normal and H OA cells

    • The OPG/RANKL ratio is significantly lower in L OA compared to normal or H OA osteoblasts (p<0.02, p<0.03)

    • Inhibition of endogenous PGE₂ production modulates these patterns

Dental pulp tissue responses:

  • Traumatic injury increases RANKL expression in pulpal fibroblastic cells while decreasing OPG

  • Damage enhances hypoxia-inducible factor-1α and -2α, which reportedly increase RANKL and decrease OPG

  • The relative RANKL/OPG ratio significantly increases in damaged pulp compared to undamaged controls

Immune cell contributions:

  • Activated T cells express RANKL that binds to RANK on dendritic cells, regulating their function and survival

  • B cells produce OPG in response to P. falciparum pRBCs stimulation in vitro, with increased production observed ex vivo in patients with malaria

This complex, tissue-specific modulation demonstrates that inflammatory regulation of the OPG/RANK/RANKL axis must be considered in a context-dependent manner when designing experimental interventions.

What are the emerging therapeutic applications of recombinant human OPG in bone and vascular disorders?

Recombinant human OPG shows promising therapeutic potential across multiple pathological conditions, with several key applications emerging from research:

Bone metabolism disorders:

  • OPG acts as a bone protector by inhibiting osteoclastogenesis through RANKL sequestration

  • The bone-protective effects make recombinant OPG a candidate for treating disorders characterized by excessive bone resorption

  • Research models suggest particular efficacy in conditions with altered OPG/RANKL ratios

Osteoarthritis interventions:

  • Paradoxical findings complicate therapeutic applications:

    • While OPG broadly inhibits bone resorption, in OA chondrocytes it significantly stimulates catabolic factors MMP-13 (p=0.05) and PAR-2 (p<0.04)

    • This suggests context-dependent effects requiring targeted delivery strategies

    • Potential for combination therapies addressing both bone and cartilage pathology

Vascular calcification prevention:

  • OPG may play a protective role against arterial calcification

  • Research links OPG to vascular pathologies including coronary artery disease, atherosclerosis, and vascular calcification

  • Therapeutic applications focus on maintaining appropriate OPG levels to prevent calcium deposition in vascular structures

Dental applications:

  • The RANKL/OPG ratio regulates odontoclastogenesis in damaged dental pulp

  • OPG expression protects pulp from odontoclastogenesis caused by traumatic damage

  • Targeted OPG delivery may help preserve dental structures following trauma

Methodological considerations for therapeutic development:

  • Delivery challenges due to the protein nature of OPG

  • Potential immunogenicity of recombinant proteins, particularly with non-human glycosylation patterns

  • Half-life considerations and dosing regimens

  • Need for tissue-specific targeting to avoid unwanted systemic effects

These emerging applications represent promising directions for translational research, leveraging insights gained from fundamental studies of OPG biology.

What are the key experimental controls needed when working with Hi-5-expressed human OPG?

Rigorous experimental design for research involving Hi-5-expressed human OPG requires comprehensive controls to ensure valid, reproducible results:

Expression and production controls:

  • Non-transfected Hi-5 cell culture supernatant as negative control

  • Parallel expression of a well-characterized control protein using the same vector and conditions

  • Monitoring protein expression kinetics (24h, 48h, 72h, 96h post-infection) to determine optimal harvest time

  • Assessment of protein stability under storage conditions (4°C, -20°C, -80°C) over different time periods

Purification validation controls:

  • Analysis of each purification fraction to track protein recovery and contaminant removal

  • Evaluation of potential His-tag interference with protein function

  • Comparison with commercially available standard OPG when possible

  • Endotoxin testing (<1 EU per 1μg of protein) to ensure preparation purity

Functional validation controls:

  • Dose-response curves for bioactivity (e.g., inhibition of RANKL-induced osteoclastogenesis)

  • Comparison with native human OPG when available

  • Heat-inactivated OPG as negative control for functional assays

  • Binding competition assays with known RANKL or TRAIL binding partners

Experimental application controls:

  • For complex cell culture experiments:

    • Include both vehicle control and irrelevant protein control

    • Test for dose-dependency across a concentration range (typically 1-100 ng/ml)

    • Include time course studies to capture dynamic responses

    • Validate results with complementary approaches (e.g., genetic knockdown of OPG receptors)

Glycosylation consideration controls:

  • Enzymatic deglycosylation to assess contribution of insect cell glycosylation to function

  • Comparison with differently glycosylated OPG variants when investigating glycan-dependent effects

Implementing these controls ensures experimental rigor and enables accurate interpretation of results when working with this recombinant protein system.

How do different recombinant protein expression systems compare for human OPG production?

Various expression systems offer distinct advantages and limitations for human OPG production, which researchers should carefully consider:

Hi-5 insect cells:

  • Yields up to 10-fold higher secreted protein compared to Sf9 cells

  • Specific protein production rate of 5.1 × 10⁻⁶ µg/(cell·h), highest among tested insect cell lines

  • Produces properly folded, bioactive OPG with appropriate disulfide bonds

  • Glycosylation pattern differs from human (high-mannose vs. complex glycans)

  • Relatively rapid production cycle (72-96 hours post-infection)

Sf9 insect cells:

  • Lower secreted protein yield compared to Hi-5 cells

  • Excellent for intracellular protein expression

  • Similar glycosylation limitations as Hi-5 cells

  • Well-established protocols and reagents available

Mammalian expression systems (CHO, HEK293):

  • Human-like complex glycosylation patterns

  • Lower protein yields compared to insect systems

  • Longer production cycles (7-14 days)

  • Higher production costs

  • More suitable for applications where human-identical glycosylation is critical

Yeast expression systems (P. pastoris):

  • High protein yields possible

  • Hyperglycosylation can occur, affecting protein properties

  • Cost-effective for scale-up

  • May require extensive optimization for proper folding of disulfide-rich proteins like OPG

Bacterial expression systems (E. coli):

  • Highest theoretical yield but often produces inclusion bodies requiring refolding

  • Lacks glycosylation capability

  • Not suitable for full-length OPG but may work for specific domains

  • Fastest production cycle (hours) and lowest cost

Comparative performance for OPG production:

Expression SystemRelative YieldGlycosylationProduction TimeCostFolding Fidelity
Hi-5 cellsVery HighHigh-mannose3-4 daysMediumHigh
Sf9 cellsHighHigh-mannose3-4 daysMediumHigh
Mammalian cellsMediumComplex7-14 daysHighVery High
YeastHighHyper3-5 daysLowMedium
E. coliLow (soluble)None1-2 daysVery LowLow

The choice of expression system should align with specific research requirements, balancing yield, glycosylation needs, timeline, and cost considerations.

What are the most sensitive detection methods for measuring OPG in biological samples?

Accurate quantification of OPG in biological samples requires sensitive, specific detection methods appropriate for research contexts:

ELISA-based methods:

  • Sandwich ELISA offers high sensitivity for human OPG detection:

    • Sensitivity: 0.07 pmol/l (≈1.4 pg/ml)

    • Standard range: 0-20 pmol/l (≈0-400 pg/ml)

    • Assay time: approximately 4.5 hours

    • Sample types: serum, plasma (EDTA, heparin, citrate)

    • Sample volume: as low as 20 μl per well

    • Precision: In-between-run CV ≤ 5%, Within-run CV ≤ 3%

Antibody considerations:

  • Capture antibody: typically polyclonal goat anti-human OPG antibody

  • Detection antibody: monoclonal mouse anti-human OPG, biotin-labeled

  • Streptavidin-HRP conjugate for signal development

PCR-based detection for expression analysis:

  • Real-time quantitative PCR for OPG mRNA measurement

  • Primer design for human OPG gene (TNFRSF11B, accession number NM_002546.3)

  • Standard PCR conditions: 95°C (10 minutes, one cycle), then 40 cycles of 95°C (10 seconds), 62°C for OPG (10 seconds), 72°C (15 seconds)

  • Relative quantification using 2^-ΔCT formula with β-actin normalization

Mass spectrometry approaches:

  • Targeted proteomics using multiple reaction monitoring (MRM)

  • Immunocapture coupled with mass spectrometry for enhanced sensitivity

  • Can detect OPG isoforms and modified forms not distinguishable by antibody-based methods

Western blotting:

  • Lower sensitivity than ELISA but provides information on molecular weight forms

  • Important for detecting proteolytic fragments or aggregates

  • Enhanced chemiluminescence detection improves sensitivity

Flow cytometry:

  • For cell-associated OPG detection

  • Allows simultaneous analysis of other markers in cellular subpopulations

Selection of the appropriate detection method depends on the research question, sample type, required sensitivity, and whether protein quantity or molecular characteristics are of primary interest.

What challenges arise when scaling up Hi-5 cell culture for increased OPG production?

Scaling up Hi-5 cell culture for enhanced OPG production presents several technical challenges requiring strategic approaches:

Cell culture optimization challenges:

  • Adaptation to suspension culture requires:

    • Gradual serum reduction protocols

    • Optimization of shaking/stirring parameters to minimize shear stress

    • Cell clumping management through media formulation and mechanical strategies

  • Scale-dependent parameters requiring adjustment:

    • Oxygen transfer rate becomes limiting in larger vessels

    • Heat transfer and temperature control requires more sophisticated systems

    • Mixing efficiency affects nutrient distribution

Process development considerations:

  • Cell density optimization:

    • Higher densities increase volumetric productivity but may impact protein quality

    • Nutrient limitations at high cell densities require feeding strategies

  • Infection parameters:

    • Multiplicity of infection (MOI) optimization is critical for yield

    • Timing of infection (cell growth phase) impacts productivity

    • Virus stability in large-scale cultures requires monitoring

Harvest and purification scale-up issues:

  • Cell separation from large volumes requires industrial centrifugation or filtration

  • Chromatography scaling considerations:

    • Column diameter to height ratio optimization

    • Flow rate adjustments to maintain residence time

    • Pressure limitations in larger columns

  • Buffer volumes increase dramatically, requiring:

    • Storage capacity planning

    • Preparation logistics

    • Cost management strategies

Quality control challenges:

  • Batch-to-batch consistency becomes more critical

  • Monitoring systems for:

    • Cell viability and growth kinetics

    • Infection efficiency

    • Protein expression levels

    • Glycosylation consistency

    • Contaminant profiles

Regulatory considerations for larger-scale production:

  • Documentation requirements increase with scale

  • Raw material traceability becomes more complex

  • Process validation requires rigorous testing

Researchers can address these challenges through systematic process development, leveraging scale-down models to predict large-scale behavior and implementing appropriate monitoring and control systems.

What emerging technologies could improve recombinant human OPG production in insect cell systems?

Several cutting-edge technologies show promise for enhancing recombinant human OPG production in insect cell systems:

Genetic engineering approaches:

  • CRISPR/Cas9 modification of Hi-5 cells to:

    • Knockout proteases that degrade secreted proteins

    • Engineer humanized glycosylation pathways

    • Enhance secretory pathway capacity

    • Integrate stable expression cassettes for continuous production

  • Promoter optimization for temporal control of expression

Bioprocess enhancements:

  • Perfusion culture systems allowing:

    • Extremely high cell densities (>10⁷ cells/mL)

    • Continuous harvest of secreted OPG

    • Extended production periods

  • Microcarrier culture in single-use bioreactors combining advantages of:

    • Adherent cell growth for protein quality

    • Suspension culture for scalability

    • Reduced risk of contamination

Expression vector innovations:

  • Development of non-lytic baculovirus expression systems

  • Design of hybrid promoters with enhanced strength and regulation

  • Self-cleaving tags for improved recombinant protein processing

  • Incorporation of molecular chaperones to enhance folding efficiency

Advanced analytical and monitoring tools:

  • Real-time monitoring of protein expression using fluorescent reporters

  • Inline protein quality assessment technologies

  • Artificial intelligence algorithms for process optimization

Cell line development:

  • Further enhancement of High Five cells through:

    • Directed evolution for increased productivity

    • Adaptation to chemically defined media

    • Selection of virus-free cell lines like Tnao38 and Tnms42

    • Development of stable cell lines with inducible expression systems

These emerging technologies, particularly when used in combination, could significantly improve both the quantity and quality of recombinant human OPG produced in insect cell systems, accelerating research applications and potential therapeutic development.

How might OPG research contribute to understanding the connection between bone metabolism and systemic inflammation?

The intersection of OPG biology with both bone metabolism and inflammation provides valuable insights into their interconnection:

Molecular crossover mechanisms:

  • OPG functions beyond bone as a mediator in multiple inflammatory contexts:

    • In malaria, OPG is increased and its ligands TRAIL and RANKL decreased in proportion to disease severity

    • OPG levels correlate with inflammatory markers like IL-6 and with endothelial activation markers including angiopoietin-2, E-selectin and ICAM-1

    • B cells have been identified as a source of OPG in response to P. falciparum infected red blood cells, suggesting immunological regulation beyond bone

Cytokine networking and regulation:

  • Inflammatory cytokines modulate the OPG/RANKL axis:

    • IL-1β, TNF-α, and PGE₂ significantly enhance both OPG and membranous RANKL levels

    • These same inflammatory mediators drive pathology in both bone and systemic inflammatory conditions

    • HIF-1α and HIF-2α, induced in hypoxic inflammatory environments, regulate RANKL/OPG expression

Clinical correlations requiring investigation:

  • Associations between inflammatory markers and bone turnover in conditions like:

    • Rheumatoid arthritis and other autoimmune disorders

    • Sepsis and critical illness

    • Chronic inflammatory states

    • Post-menopausal bone loss with low-grade inflammation

Therapeutic implications:

  • Targeted modulation of OPG/RANKL might address both inflammatory and skeletal manifestations

  • Anti-inflammatory approaches may have indirect benefits on bone metabolism through OPG/RANKL regulation

  • Combined therapeutic strategies addressing both systems simultaneously

Product Science Overview

Introduction

Osteoprotegerin (OPG), also known as tumor necrosis factor receptor superfamily member 11B (TNFRSF11B), is a crucial protein involved in bone remodeling and homeostasis. It acts as a decoy receptor for receptor activator of nuclear factor kappa B ligand (RANKL), thereby inhibiting osteoclastogenesis and promoting osteoclast apoptosis .

Discovery and Nomenclature

OPG was independently discovered by two research teams in 1997. Boyle’s group identified it while generating transgenic mice overexpressing various TNF receptor cDNAs, and Tsuda’s group identified it as a repressor of osteoclast differentiation . The protein was named osteoprotegerin from the Latin words “osteo” (bone) and “protegere” (to protect) .

Biological Function

OPG plays a central role in bone remodeling, a biological mechanism where around 10% of total bone mass is renewed per year . This process involves a balance between osteoclasts, which resorb bone, and osteoblasts, which form new bone. OPG inhibits the activation of osteoclasts by neutralizing RANKL, thus preventing excessive bone resorption .

Recombinant OPG (Human Recombinant, Hi-5)

Human recombinant OPG, particularly the Hi-5 variant, is produced using advanced biotechnological methods. This recombinant form retains the biological activity of natural OPG and is used in various research and therapeutic applications . It is often tagged with a His-tag to facilitate purification and detection .

Clinical and Research Applications

OPG has been extensively studied for its role in bone diseases such as osteoporosis and bone metastasis in cancer . It is also being explored as a potential therapeutic target for various cancers due to its involvement in tumor survival, epithelial to mesenchymal transition (EMT), neo-angiogenesis, invasion, and metastasis .

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.