NOV Human, HEK

Nephroblastoma Overexpressed Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Overview of NOV Human, HEK

NOV Human, HEK refers to a recombinant form of the Nephroblastoma Overexpressed (NOV) protein, also known as CCN3, produced in Human Embryonic Kidney 293 (HEK293) cells. This glycoprotein belongs to the CCN family of matricellular proteins, which regulate cellular processes such as adhesion, proliferation, and differentiation. The HEK293 expression system ensures proper post-translational modifications (PTMs), critical for the protein’s structural and functional integrity .

Expression System

HEK293 cells are ideal for NOV production due to:

  • High transfection efficiency: Enables robust recombinant protein yields .

  • Human-like PTMs: Ensures proper γ-carboxylation and sulfation absent in CHO cells .

  • Scalability: Adaptability to serum-free suspension cultures for industrial-scale production .

Purification and Stability

  • Lyophilized NOV retains stability for long-term storage at -80°C.

  • Reconstitution requires filtration (0.4 µm) to remove aggregates .

Functional Applications

NOV Human, HEK is utilized in:

  • Cancer research: Studying its role in nephroblastoma and other tumors via cell adhesion assays .

  • Tissue regeneration: Investigating modulation of extracellular matrix interactions .

  • Drug discovery: Screening for agonists/antagonists targeting NOV signaling pathways .

Comparative Performance

FeatureHEK293CHO Cells
Glycosylation ProfileHuman-like, no Neu5Gc/α-GalNon-human glycans
γ-Carboxylation EfficiencyHighLow
Tyrosine SulfationRobustLimited
Regulatory ApprovalFDA/EMA-approved therapeuticsDominant in industry

Limitations

  • Lower volumetric yields compared to CHO cells .

  • Batch variability due to endogenous ion channel activity (e.g., NaV1.7) .

Research Findings and Clinical Relevance

  • HEK293-derived NOV lacks immunogenic non-human epitopes, making it suitable for in vivo studies .

  • HEK293 systems have produced FDA-approved biologics (e.g., NUWIQ®, a recombinant Factor VIII), validating their clinical utility .

Future Directions

  • CRISPR engineering: Enhancing HEK293 productivity via GLUL knockout or metabolic pathway optimization .

  • Single-use bioreactors: Scaling NOV production for therapeutic applications .

Product Specs

Introduction

Nephroblastoma Overexpressed (NOV), encoded by the NOV gene, belongs to the CCN (CTGF/CYR61/NOV) family. NOV plays a role in suppressing tumor development and the rapid multiplication of cells in certain cancer cell lines. It interacts with various proteins and participates in both intracellular and extracellular signaling pathways. NOV is found in specific tumors, including Wilms' tumor and most nephroblastomas. Additionally, it exhibits proangiogenic properties, promoting the formation of new blood vessels.

Description

This product consists of human recombinant NOV protein produced in HEK293 cells. It is a single, glycosylated polypeptide chain with a molecular weight of 36.5kDa (calculated). The protein sequence spans amino acids 33 to 357 and includes a 6-amino acid C-terminal His tag.

Physical Appearance
The product appears as a white powder that has been freeze-dried and subjected to filtration.
Formulation

The NOV protein has undergone filtration (0.4 µm) and lyophilization. The initial concentration before lyophilization was 0.5mg/ml in a solution of PBS with 5% (w/v) trehalose.

Solubility

To prepare a working stock solution, it is advised to add deionized water to the lyophilized pellet until it fully dissolves, achieving an approximate concentration of 0.5mg/ml. Please note that this product is not sterile. Before using it in cell culture, it is essential to filter the solution through an appropriate sterile filter.

Stability

For long-term storage, the lyophilized protein should be kept at -20°C. After reconstituting the protein, it is recommended to aliquot it into smaller portions to minimize repeated freezing and thawing cycles. The reconstituted protein can be stored at 4°C for a limited duration, maintaining stability for up to one week.

Purity

Analysis by SDS-PAGE indicates that the purity of this product is greater than 90.0%.

Synonyms

Protein NOV homolog, NovH, CCN family member 3, nsulin-like growth factor-binding protein 9, IBP-9, IGF-binding protein 9, IGFBP-9, Nephroblastoma-overexpressed gene protein homolog, NOV, CCN3, IGFBP9, NOVH.

Source

HEK293 cells.

Amino Acid Sequence

QRCPPQCPGR CPATPPTCAP GVRAVLDGCS CCLVCARQRG ESCSDLEPCD ESSGLYCDRS ADPSNQTGIC TAVEGDNCVF DGVIYRSGEK FQPSCKFQCT CRDGQIGCVP RCQLDVLLPE PNCPAPRKVE VPGECCEKWI CGPDEEDSLG GLTLAAYRPE ATLGVEVSDS SVNCIEQTTE WTACSKSCGM GFSTRVTNRN RQCEMLKQTR LCMVRPCEQE PEQPTDKKGK KCLRTKKSLK AIHLQFKNCT SLHTYKPRFC GVCSDGRCCT PHNTKTIQAE FQCSPGQIVK KPVMVIGTCT CHTNCPKNNE AFLQELELKT TRGKMHHHHH H.

 

Q&A

What are HEK293 cells and why are they significant for protein expression research?

HEK293 cells are a human embryonic kidney cell line that has become one of the most widely used mammalian expression systems in biomedical research. Their significance stems from several key characteristics:

  • Human origin providing authentic human post-translational modifications

  • Exceptional efficiency in complex modifications including glutamic acid γ-carboxylation and tyrosine sulfation

  • Established regulatory track record with several approved therapeutic proteins

  • Adaptability to serum-free suspension cultures for large-scale applications

Several protein therapeutics produced in HEK293 cells, including recombinant factor VIII-Fc, Dulaglutide, Idursulfase, and Velaglucerase alfa, have already received FDA and EMA approval, demonstrating their suitability for therapeutic protein production .

How do the glycosylation capabilities of HEK293 cells compare with non-human expression systems?

HEK293 cells offer significant advantages in glycosylation compared to non-human expression systems:

FeatureHEK293 CellsCHO/Non-Human CellsImpact on Research
Non-human glycansAbsent (no α-Gal or Neu5Gc)Present (Neu5Gc found in 4.7% of CHO glycopeptides)Eliminates immunogenic risk
SialylationHuman sialic acid (Neu5Ac)Can produce non-human Neu5GcAuthentic human patterns
Specific enzymesExpresses human-specific glycosyltransferasesLacks GnT-III, Gal α2,6 ST, α1,3/4 fucosyltransferaseMore complex human glycoforms
LacdiNAc structuresPresentTypically absentHuman-like glycan diversity

Mass spectrometry analysis has demonstrated that HEK293-produced erythropoietin (EPO) contains no alpha-Gal or Neu5Gc epitopes, while these non-human glycans are detected in CHO-derived EPO . These non-human glycosylations in CHO and other mammalian cell lines raise the possibility of immunological responses to biotherapeutics .

What are the key considerations when selecting between HEK293 and other human cell lines for research?

When choosing between HEK293 and other human cell lines for research applications, consider:

  • Regulatory Status: HEK293 and HT-1080 have established regulatory track records with approved therapeutics, while AGE1.HN, CAP, HKB-11, and PER.C6 products are still in various developmental phases .

  • Specific Modifications: HEK293 cells are "exceptionally efficient in glutamic acid γ-carboxylation and tyrosine sulfation," critical for certain proteins like coagulation factors .

  • Growth Characteristics: Different HEK293 variants (HEK293T, HEK293-EBNA) offer specialized features like improved transfection efficiency or protein production capacity .

  • Experimental Goals: For signaling studies, HEK293T cells provide excellent transfection efficiency for reporter assays, while stable protein production might benefit from specialized clones .

  • Genetic Background: Consider that HEK293 cells contain approximately 112 unique variants identified at the proteome level, including variants in cancer-related proteins like p53 .

What genetic engineering strategies can enhance recombinant protein production in HEK293 cells?

Multiple genetic engineering approaches have been developed to maximize protein expression in HEK293 systems:

  • GLUL Knockout System: A particularly effective approach involves:

    • CRISPR-Cas9 knockout of endogenous GLUL (glutamine synthetase) gene

    • Introduction of expression vectors using human GLUL as selection marker

    • Selection with methionine sulfoximine (MSX) to identify high-expressing cells

  • Gene Copy Number Optimization: Digital PCR analysis of high-producing HEK293 cells shows:

    • Successful cell pools contain approximately four-fold higher exogenous GLUL copy number compared to endogenous GLUL in wildtype cells

    • Similar four-fold increase in target protein gene copies (e.g., EPO)

  • Vector Design Considerations:

    • Selection marker and target gene expression must be carefully balanced

    • Promoter strength and regulatory elements significantly impact expression levels

    • Integration site effects can be mitigated through specialized vector designs

This engineered approach has yielded impressive results, with documented EPO production levels reaching 92,700 U/mL (or 696 mg/L) in 2L stirred-tank fed-batch bioreactors .

How can researchers optimize glycosylation profiles of proteins expressed in HEK293 cells?

Researchers can control glycosylation profiles through several methodological approaches:

  • Site-Specific Glycosylation Analysis:

    • Mass spectrometry enables detailed mapping of N-glycosylation and O-glycosylation patterns

    • Analysis of HEK293-expressed EPO revealed site-specific differences in sialylation (Site 3 > Site 2 > Site 1)

    • Quantitative assessment demonstrated complete N-glycosylation at Sites 1 and 2, with 99.96% occupancy at Site 3

  • Metabolic Engineering Strategies:

    • Media supplementation with specific glycan precursors

    • Introduction of additional glycosyltransferases for desired structures

    • Regulation of sialyltransferase activity to control terminal sialylation

  • Process Parameter Optimization:

    • Temperature, pH, and dissolved oxygen affect glycosylation patterns

    • Nutrient feeding strategies impact glycan processing enzymes

    • Harvesting time influences terminal glycan processing

The glycosylation profile of HEK293-produced EPO shows complete core fucosylation (2.98 moles out of possible 3.0 moles), high sialylation (average 6.55 NeuAc per mole), and the presence of human-specific structures like LacdiNAc, which may enhance galectin-3 binding and potentially improve pharmacokinetics .

What bioreactor parameters are critical for scaling up HEK293 cell cultures?

Scaling up HEK293 cultures requires careful control of key bioreactor parameters:

ParameterOptimal RangeImpact on ProductionMonitoring Approach
Dissolved oxygen30-50% saturationAffects metabolism and glycosylationReal-time DO probes
pH7.0-7.2Influences growth and protein qualityAutomated pH control
Temperature37°C standard; 32-34°C for production phaseRegulates growth vs. production balancePrecise temperature control
AgitationSystem-dependent (60-150 rpm)Balances mixing with shear stressPower input measurement
Feeding strategyProcess-specificMaintains nutrients and removes wasteMetabolite analysis
Cell densityTarget 5-15×10^6 cells/mLOptimizes volumetric productivityAutomated cell counting

Successful scale-up has been demonstrated with HEK293 cells producing EPO at 696 mg/L in 2L stirred-tank fed-batch bioreactors . This indicates that with proper parameter optimization, HEK293 cells can achieve production levels suitable for research and potential therapeutic applications.

How can HEK293 cells be utilized for studying cell signaling pathways?

HEK293 cells serve as versatile tools for dissecting complex signaling pathways:

  • Reporter Assay Systems:

    • Transfection with pathway-responsive elements (e.g., pTOPFLASH/pFOPFLASH for Wnt/β-catenin)

    • Measurement of luciferase activity in response to pathway activation/inhibition

    • Quantitative assessment of signaling modulation by candidate genes

  • Protein Interaction Studies:

    • Co-immunoprecipitation with tagged proteins (e.g., FLAG-tagged β-catenin, GSK3-β)

    • Visualization of protein complexes through fluorescent tagging

    • Functional assessment of interaction through mutational analysis

  • Pathway Component Manipulation:

    • Expression of wild-type vs. mutant pathway components

    • siRNA knockdown of target genes (e.g., MOCA knockdown)

    • CRISPR-Cas9 editing of pathway regulators

In a specific example from the search results, HEK293T cells were used to investigate how MOCA (modifier of cell adhesion) regulates Wnt/β-catenin signaling. Researchers demonstrated that "MOCA forms a complex with β-catenin and inhibits transcription of known Wnt target genes" .

What approaches can researchers use to establish stable HEK293 cell lines?

Creating stable HEK293 cell lines requires a systematic approach:

  • Selection System Design:

    • CRISPR-Cas9 knockout of endogenous selection markers (e.g., GLUL)

    • Transfection with vectors containing both selection marker and target gene

    • Stringent selection with appropriate agents (e.g., MSX for GLUL system)

  • Clonal Isolation Methods:

    • Limiting dilution for single-cell isolation

    • FACS sorting based on reporter gene expression

    • Automated single-cell deposition systems

  • Stability Assessment:

    • Gene copy number verification via droplet digital PCR (ddPCR)

    • Analysis shows stable cell pools maintain ~4-fold higher exogenous gene copies

    • Product quality consistency assessment over extended passages

  • Productivity Screening:

    • High-throughput analytical methods for protein quantification

    • Functional assays for bioactivity assessment

    • Glycosylation and other PTM characterization

Researchers have documented that stable HEK293 cell pools selected using the GLUL/MSX system can maintain high productivity, achieving EPO production levels of 92,700 U/mL as measured by ELISA or 696 mg/L by densitometry in bioreactor cultures .

How can proteogenomic approaches enhance our understanding of HEK293 cell biology?

Proteogenomic analysis provides valuable insights into HEK293 cell biology:

  • Variant Identification Strategy:

    • Integration of exome sequencing data with shotgun proteome analysis

    • Custom database generation incorporating identified genomic variants

    • Mass spectrometry validation of variant peptides

  • Key Findings from HEK293 Proteogenomics:

    • 112 unique variants identified at the proteome level out of ~1200 coding variants in the exome

    • 7 variants shared across multiple proteomic datasets

    • At least 8 proteins with variants were annotated as cancer-related, including p53 tumor suppressor

  • Technical Considerations:

    • Variant peptides were 2.5 times less likely to be identified than wild-type peptides

    • This may result from the presence of "passenger" mutations in genes

    • Multiple complementary proteomic approaches improve variant detection

  • Research Applications:

    • Assessment of how genetic variations affect protein function

    • Understanding cellular adaptation mechanisms

    • Evaluation of how variants might impact experimental outcomes

Proteogenomic analysis helps researchers understand the unique genetic and proteomic landscape of HEK293 cells, providing context for experimental design and interpretation.

What are the advantages and limitations of HEK293 versus CHO cells for different research applications?

A systematic comparison reveals distinct advantages and limitations:

CriterionHEK293 AdvantageCHO AdvantageResearch Implications
GlycosylationHuman-like patterns, no immunogenic glycansHigher batch consistencyHEK293 superior for studying human glycoproteins
Complex PTMsEfficient γ-carboxylation, tyrosine sulfationLess complex but more consistent PTMsHEK293 better for complex human proteins
ProductivityImproving with new engineering approachesHistorically higher titersApplication-dependent selection
Regulatory KnowledgeGrowing but still limitedExtensive regulatory historyCHO preferable for straightforward therapeutics
Specific ApplicationsHuman neurobiology, complex signalingSimple protein expression, antibodiesChoose based on research question

Mass spectrometry analysis reveals that "N-glycosylation of the produced EPO was similar to endogenous human proteins and non-human glycan epitopes were not detected" in HEK293 cells, while CHO-derived EPO contained Neu5Gc in 4.7% of glycopeptide spectra . This makes HEK293 particularly valuable for studying proteins where human-authentic glycosylation is critical.

For which specific protein classes are HEK293 cells demonstrably superior to alternative expression systems?

HEK293 cells show clear advantages for specific protein classes:

  • Coagulation Factors:

    • Superior in glutamic acid γ-carboxylation

    • Critical for proteins like Drotrecogin alfa and recombinant factor IX-Fc

    • Enables proper folding and functional activity

  • Complex Glycoproteins:

    • Elimination of immunogenic non-human glycans

    • Authentic human N-glycan and O-glycan structures

    • Particularly valuable for erythropoietin and other cytokines

  • Multi-domain Human Proteins:

    • Proper folding and assembly of complex structures

    • Appropriate disulfide bond formation

    • Correct proteolytic processing

  • Cell Surface Receptors:

    • Appropriate trafficking and membrane insertion

    • Human-like glycosylation affecting ligand binding

    • Useful for studying receptor function (e.g., Frizzled receptors)

  • Signaling Proteins:

    • Correct post-translational modifications affecting activity

    • Proper binding to human interaction partners

    • Authentic regulation in signaling pathways

FDA and EMA approvals for therapeutics from HEK293 cells, including "recombinant factor VIII-Fc, Dulaglutide, Idursulfase and Velaglucerase alfa" demonstrate their advantage for complex human proteins .

How do recent advances in gene editing technologies impact the comparative advantages of expression systems?

Gene editing technologies are reshaping the landscape of expression systems:

  • CRISPR-Cas9 Applications in HEK293:

    • Precise knockout of endogenous genes (e.g., GLUL)

    • Creation of specialized selection systems

    • Enhancement of specific cellular functions

  • Humanization of Non-human Systems:

    • Genetic modification of CHO cells to produce more human-like glycans

    • Introduction of human-specific enzymes in non-human cells

    • Gap between HEK293 and modified CHO narrowing for certain applications

  • Engineering HEK293 for Enhanced Performance:

    • Knockout of proteases that degrade recombinant proteins

    • Introduction of productivity-enhancing genes

    • Modification of metabolic pathways for improved growth

  • Methodological Considerations:

    • CRISPR-Cas9 efficiency in HEK293 typically exceeds that in CHO

    • Multiple genetic modifications more feasible in well-characterized systems

    • Off-target effects must be carefully assessed in all systems

The CRISPR-Cas9 system has been successfully used to knock out GLUL in HEK293 cells, creating a platform for developing high-producing cell lines through MSX selection of cells transfected with GLUL-based expression vectors . This demonstrates how gene editing can dramatically enhance the utility of human cell expression systems.

How might advances in synthetic biology reshape HEK293-based expression systems?

Synthetic biology approaches offer transformative potential for HEK293 expression systems:

  • Genome Minimization:

    • Removal of non-essential genes to create streamlined cells

    • Elimination of endogenous retroviral elements

    • Development of "clean" genetic backgrounds for specialized applications

  • Orthogonal Translation Systems:

    • Integration of synthetic amino acid incorporation machinery

    • Production of proteins with non-canonical amino acids

    • Novel functionalization possibilities for research tools

  • Synthetic Regulatory Circuits:

    • Programmable gene expression systems

    • Feedback-controlled production based on cellular states

    • Temporal control of expression for complex protein assemblies

  • Engineering Cellular Compartments:

    • Optimization of secretory pathway components

    • Enhancement of post-translational modification machinery

    • Creation of specialized microenvironments for protein folding

These approaches, while not specifically discussed in the search results, represent logical extensions of current HEK293 engineering efforts such as the GLUL knockout system that has already demonstrated substantial productivity improvements .

What research challenges remain in understanding the full capabilities of HEK293 cells?

Several key challenges continue to shape HEK293 research:

  • Genetic Heterogeneity:

    • Proteogenomic analysis has identified 112 unique variants at the protein level

    • Impact of these variants on cellular function requires further investigation

    • At least 8 cancer-related proteins show variations, including p53 tumor suppressor

  • Glycosylation Complexity:

    • Full understanding of site-specific glycosylation patterns

    • Functional significance of structures like LacdiNAc in HEK293-produced proteins

    • Potential interactions with human galectin-3 and implications for protein half-life

  • Cell Line Evolution:

    • Long-term genetic stability assessment

    • Epigenetic changes during extended culture

    • Comparison of different HEK293 derivatives (HEK293T, HEK293-EBNA, etc.)

  • Functional Genomics:

    • Comprehensive mapping of HEK293 cellular pathways

    • Understanding of endogenous regulatory networks

    • Cell type-specific responses to environmental factors

Addressing these challenges will enhance our ability to leverage HEK293 cells for increasingly sophisticated research applications and potentially expand their utility in therapeutic protein production.

How might the growing acceptance of human cell lines impact regulatory considerations for research applications?

The increasing adoption of human cell lines has important regulatory implications:

  • Evolving Regulatory Landscape:

    • Growing acceptance of human cell-derived therapeutics

    • Establishment of specialized guidelines for human cell lines

    • Shift in risk assessment frameworks

  • Biosafety Considerations:

    • Previous concerns about "potential biosafety risks from human virus contamination"

    • Recent regulatory approvals demonstrate "successful circumvention" of these concerns

    • Development of enhanced screening protocols

  • Standardization Efforts:

    • Creation of well-characterized human cell banks

    • Development of reference standards for human cell-derived proteins

    • Harmonization of testing requirements across regulatory jurisdictions

  • Ethical Considerations:

    • Transparent sourcing and documentation of human cell lines

    • Informed consent frameworks for primary cell derivation

    • Ethical guidelines for genetic modification of human cells

The regulatory track record established by approved HEK293-derived therapeutics provides "a useful basis for future assessment of other therapeutics produced using human cell lines" , suggesting continued expansion of human expression systems in both research and therapeutic applications.

Product Science Overview

Introduction

Nephroblastoma Overexpressed (NOV), also known as CCN3, is a member of the CCN family of proteins. This family includes connective tissue growth factor (CTGF/CCN2), cysteine-rich angiogenic inducer 61 (CYR61/CCN1), and nephroblastoma overexpressed (NOV/CCN3). These proteins are involved in various cellular processes, including cell adhesion, migration, proliferation, and differentiation .

Gene and Protein Structure

The NOV gene encodes a matricellular protein that is involved in the regulation of cell-matrix interactions. The protein structure of NOV includes several conserved domains: an insulin-like growth factor-binding protein (IGFBP) domain, a von Willebrand factor type C (vWC) domain, a thrombospondin type 1 (TSP1) domain, and a cysteine knot (CT) domain. These domains are crucial for the protein’s interaction with other cellular components and its role in cellular signaling pathways .

Biological Functions

NOV/CCN3 has been shown to have growth-inhibiting properties. It plays a role in regulating extracellular matrix protein expression and influences its own expression when overexpressed. The protein is involved in various biological processes, including tissue repair, fibrosis, and tumorigenesis. NOV/CCN3 acts as a downstream mediator of transforming growth factor-beta 1 (TGF-β1) signaling, which is involved in tissue scarring and the stimulation of extracellular matrix protein deposition .

Recombinant Expression in HEK Cells

Human embryonic kidney (HEK) cells are commonly used for the recombinant expression of proteins due to their high transfection efficiency and ability to perform post-translational modifications. In the case of NOV/CCN3, stable HEK cell lines have been established to produce recombinant human NOV/CCN3 protein. These cell lines are used to generate large quantities of the protein for research and therapeutic purposes .

Purification and Characterization

The recombinant NOV/CCN3 protein is purified using various chromatographic techniques. The identity of the purified protein is confirmed through in-gel digestion followed by mass spectrometry. Functional characterization of the protein is performed using assays such as the Smad3-sensitive reporter gene assay and the BrdU proliferation assay. These assays help determine the biological activity of the recombinant protein .

Applications and Future Directions

The recombinant NOV/CCN3 protein has potential applications in the diagnosis and treatment of fibrotic diseases and cancer. Its role in regulating extracellular matrix protein expression makes it a promising target for therapeutic interventions. Further research is needed to fully understand the mechanisms by which NOV/CCN3 influences cellular processes and to develop effective therapies based on this protein .

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.