Recombinant Macaca mulatta TNF protein (partial, active) is a bioengineered cytokine derived from the extracellular domain (ECD) of tumor necrosis factor-alpha (TNF-α). Engineered in E. coli, this protein is designed for research applications in primate immunology, inflammation, and disease modeling. It retains 97% amino acid sequence identity with human TNF-α , enabling cross-species functional studies in rhesus macaque models.
Storage: -20°C (stable for >12 months) ; avoid repeated freeze-thaw cycles .
Reconstitution: 100 µg/mL in PBS with BSA (if carrier-free, use sterile PBS) .
Assay | Result |
---|---|
ED₅₀ (L929 cells + actinomycin D) | <0.05 ng/mL |
Specific Activity | >2.0 × 10⁷ IU/mg |
Receptor Affinity | Binds TNFR1 and TNFR2 with comparable affinity |
The protein induces apoptosis in murine L929 cells at nanomolar concentrations, confirming its bioactivity . Its trimeric structure enables interaction with TNF receptors, triggering NF-κB and MAPK signaling pathways .
Primate Immunology:
In Vitro Cytotoxicity:
Cross-Species Reactivity:
TNF-alpha, also known as cachectin and TNFSF2, is a pleiotropic molecule that plays central roles in inflammation, immune system development, apoptosis, and lipid metabolism . In rhesus macaques, TNF-alpha consists of a 35 amino acid cytoplasmic domain, a 21 amino acid transmembrane segment, and a 177 amino acid extracellular domain (ECD) . It functions as a key mediator of inflammatory responses, similar to human TNF-alpha.
TNF-alpha is produced by various immune cells (macrophages, monocytes, T cells, B cells), as well as epithelial cells, endothelial cells, and tumor cells . The protein is assembled intracellularly to form a noncovalently linked homotrimer expressed on the cell surface, which can induce lysis of neighboring tumor cells and virus-infected cells . Membrane-bound TNF-alpha can be cleaved by TACE/ADAM17 to release a bioactive soluble cytokine .
The biological effects of TNF-alpha are mediated through two receptors: the ubiquitous 55-60 kDa TNF RI, which contains a cytoplasmic death domain triggering apoptosis, and the hematopoietic cell-restricted 80 kDa TNF RII . Both receptors bind TNF-alpha with comparable affinity .
Rhesus macaque TNF-alpha shares remarkable homology with human TNF-alpha, making it valuable for translational research. Within the extracellular domain, rhesus TNF-alpha shares 97% amino acid sequence identity with human TNF-alpha . This high conservation extends to the structural organization of the protein, with both species having similar domain structures.
Despite this similarity in protein sequence, there are important differences in the regulatory regions. Polymorphisms in the human TNF-alpha promoter known to be associated with malaria susceptibility are not shared with macaques . Sequence analysis of the TNF-alpha promoter region in macaques revealed a total of 14 single nucleotide polymorphisms (SNPs), with 20 unique haplotypes identified across different macaque populations .
Recombinant rhesus macaque TNF-alpha is typically produced as the soluble extracellular domain portion of the native protein, spanning from Val77 to Leu233 . Based on comparative analysis with human TNF-alpha, we can infer that rhesus TNF-alpha forms a homotrimer with a molecular weight of approximately 53 kDa in its native state .
When analyzed by SDS-PAGE under reducing conditions, the monomeric form appears as a band at approximately 17 kDa . The trimeric structure is critical for biological activity, as it enables proper receptor binding and signaling.
E. coli-derived rhesus macaque TNF-alpha is commonly used in research applications . The recombinant protein can be produced either with a carrier protein (typically bovine serum albumin, BSA) to enhance stability or in carrier-free formulations for applications where BSA might interfere .
Evaluating the biological activity of recombinant rhesus macaque TNF-alpha requires well-designed assays that capture its key functions. The most established method is the L-929 mouse fibroblast cytotoxicity assay in the presence of actinomycin D. The ED50 (effective dose for 50% cytotoxicity) for biologically active rhesus macaque TNF-alpha should be in the range of 15-60 pg/mL .
For this assay:
Seed L-929 cells in 96-well plates
Add actinomycin D (typically 1 μg/mL)
Add serial dilutions of TNF-alpha
Assess cell viability after 18-24 hours
Calculate ED50 values
Additional assays to consider include:
NF-κB activation assays: Using reporter cell lines expressing luciferase under NF-κB responsive elements to measure signaling pathway activation
Proliferation assays: Recombinant TNF-alpha can enhance proliferation of certain cell types, such as rhesus B cell lines
Cytokine induction: Measure the production of downstream inflammatory cytokines (IL-1β, IL-6, IL-8) in response to TNF-alpha stimulation
Western blotting: To detect phosphorylation of signaling molecules in the TNF pathway (p38 MAPK, JNK, IKK)
Always include appropriate controls:
Positive controls: Human TNF-alpha
Negative controls: Heat-inactivated TNF-alpha, unrelated proteins
Specificity controls: TNF-alpha neutralizing antibodies
Genetic polymorphisms in the TNF-alpha promoter region can significantly impact TNF-alpha expression in rhesus macaques, potentially influencing disease susceptibility and experimental outcomes. A study examining 40 macaques, including M. mulatta of Chinese and Indian ancestry and M. fascicularis, identified important genetic variations .
The analysis revealed 14 single nucleotide polymorphisms (SNPs) in the TNF-alpha promoter region, five of which were newly described at the time . These polymorphisms can potentially influence transcription factor binding, affecting regulation of TNF-alpha expression. The TFSEARCH program was used to investigate the potential of these polymorphisms to influence transcription factor binding .
These genetic variations could contribute to differential disease susceptibility observed among different macaque populations. Researchers should consider the genetic background of animals used in their studies, as differences in TNF-alpha promoter polymorphisms could affect experimental results and their interpretation.
Proper storage and handling of recombinant rhesus macaque TNF-alpha is critical for maintaining its biological activity. Based on product information, the following conditions are recommended:
For lyophilized protein:
Reconstitution protocols:
For preparations with BSA carrier: Reconstitute at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin
For carrier-free preparations: Reconstitute at 100 μg/mL in sterile PBS without additives
Storage after reconstitution:
Prepare aliquots to avoid repeated freeze-thaw cycles
Store reconstituted protein at -20°C to -80°C
Use a manual defrost freezer to avoid temperature fluctuations
Avoid exposure to strong light and oxidizing agents
Handling precautions:
Thaw aliquots on ice or at 2-8°C, never at room temperature
Once thawed, use immediately or store at 2-8°C for short-term (1-2 weeks)
Prepare working dilutions on ice and use within the same day
The presence of carrier proteins like BSA enhances protein stability and increases shelf-life . For applications where BSA could interfere (such as certain immunoassays), use carrier-free preparations.
Comprehensive characterization of recombinant rhesus macaque TNF-alpha requires multiple analytical approaches to assess identity, purity, structure, and biological activity:
Physicochemical Characterization:
SDS-PAGE Analysis: Under reducing conditions, monomeric TNF-alpha appears as a band at approximately 17 kDa . Silver staining provides higher sensitivity for detecting impurities.
Size Exclusion Chromatography (SEC): For analyzing the oligomeric state (homotrimer) and aggregation status. SEC coupled with multi-angle light scattering (SEC-MALS) can determine absolute molecular weight, which should be approximately 53 kDa for the trimeric form .
Mass Spectrometry: For precise molecular weight determination and primary structure confirmation.
Immunological Characterization:
ELISA: For quantitative determination of concentration.
Western Blotting: For identity confirmation using specific antibodies.
Surface Plasmon Resonance (SPR): For measuring binding kinetics to TNF receptors.
Biological Activity Assessment:
Cytotoxicity Assay: The L-929 mouse fibroblast cytotoxicity assay with actinomycin D. The ED50 should be in the range of 15-60 pg/mL .
Cell-Based Reporter Assays: Such as NF-κB reporter assays to measure activation of downstream signaling.
Proliferation Assays: To assess the ability to enhance B cell proliferation .
Contaminant Analysis:
Endotoxin Testing: Using Limulus Amebocyte Lysate (LAL) assay, critical for E. coli-derived proteins.
Host Cell Protein (HCP) Analysis: Using ELISA or mass spectrometry-based methods.
By combining these methods, researchers can thoroughly characterize recombinant rhesus macaque TNF-alpha for research applications.
Determining the optimal concentration of recombinant rhesus macaque TNF-alpha requires systematic titration based on assay type:
Step 1: Literature-based starting point
From published data, we know that for cytotoxicity assays in L-929 cells with actinomycin D, the ED50 is 15-60 pg/mL . This provides a reference range for initial testing.
Prepare serial dilutions spanning 5-6 log concentrations (e.g., 0.1 pg/mL to 10 μg/mL)
Include vehicle control (0 pg/mL)
Assess biological response using appropriate readouts
Plot the dose-response curve and identify:
Minimum effective concentration
ED50 (concentration producing 50% of maximum response)
Saturation concentration
Step 3: Refined titration
Narrow the concentration range around the effective zone identified in Step 2, using smaller increments.
Concentration guidelines for different assay types:
Additional considerations:
Time-dependence: Test multiple time points to capture optimal response
Cell density: Optimize cell numbers for each assay type
Medium composition: Serum can contain TNF binding proteins
Species specificity: Rhesus TNF-alpha may have different potency on cells from different species
This systematic approach will help determine the appropriate concentration range specific to your experimental system.
Detecting TNF-alpha expression in rhesus macaque tissue samples requires methods that are both sensitive and specific:
RNA-Based Detection Methods:
RT-PCR and qRT-PCR:
Design primers specific to rhesus macaque TNF-alpha sequence
Target regions that differ from closely related species
Include appropriate housekeeping genes for normalization
Provides quantitative assessment of TNF-alpha mRNA levels
RNA In Situ Hybridization (RNA-ISH):
Allows visualization of TNF-alpha mRNA within tissue context
Preserves spatial information about cell types expressing TNF-alpha
Can be combined with immunohistochemistry for cell type identification
Protein-Based Detection Methods:
ELISA:
For quantitative measurement of TNF-alpha protein in tissue homogenates
Use commercially available kits validated for rhesus macaque samples
Typical detection limit: 1-10 pg/mL
Western Blotting:
Can distinguish between membrane-bound (26 kDa) and soluble (17 kDa) forms
Requires validation of antibody specificity for rhesus macaque TNF-alpha
Immunohistochemistry (IHC):
Preserves tissue architecture and cellular context
Use antibodies validated for rhesus macaque TNF-alpha
Consider antigen retrieval methods to improve detection
Flow Cytometry:
For detecting TNF-alpha in single-cell suspensions
Particularly useful for identifying specific cell populations producing TNF-alpha
Requires tissue disaggregation and intracellular staining protocols
Optimization considerations:
Method | Sample Preparation | Key Optimization Factors | Typical Sensitivity |
---|---|---|---|
qRT-PCR | RNA extraction | Primer efficiency, reference gene selection | 10-100 copies |
ELISA | Tissue homogenization | Antibody specificity, sample dilution | 1-10 pg/mL |
IHC | Fixation, sectioning | Antigen retrieval, antibody titration | Cell-level detection |
Flow Cytometry | Cell isolation | Stimulation conditions, fluorophore selection | Cell-level detection |
Combining multiple detection methods provides the most comprehensive assessment of TNF-alpha expression in tissue samples.
When designing experiments with recombinant rhesus macaque TNF-alpha, proper controls are essential for valid interpretation of results:
For in vitro studies:
Negative Controls:
Vehicle Control: The same buffer used for TNF-alpha dilution (e.g., PBS with carrier protein)
Heat-Inactivated TNF-alpha: TNF-alpha that has been heat-denatured (e.g., 95°C for 10 minutes)
Unrelated Recombinant Protein: Another recombinant protein of similar size prepared using the same expression system
Unstimulated Cells: Cells maintained in culture media without any treatment
Positive Controls:
Human TNF-alpha: Given the 97% sequence identity, this can serve as a reference standard
Known TNF-alpha Inducers: For instance, LPS for macrophage activation
Established Cell Lines: Cell lines with well-characterized responses to TNF-alpha
Specificity Controls:
TNF-alpha Neutralizing Antibodies: To confirm that observed effects are specifically due to TNF-alpha
TNF Receptor Antagonists: To confirm receptor-mediated effects
Dose-Response: Multiple concentrations of TNF-alpha to establish relationship between dose and response
For in vivo studies:
Study Design Controls:
Vehicle Treatment: Animals receiving the same vehicle used for TNF-alpha administration
Irrelevant Protein Control: Animals receiving an unrelated recombinant protein
TNF-alpha Blockade: Co-administration of TNF-alpha with neutralizing antibodies
Dose-Response Studies: Multiple dosage groups
Time-Course Studies: Sampling at multiple time points
Incorporating these controls helps attribute observed effects specifically to recombinant rhesus macaque TNF-alpha and distinguishes them from experimental artifacts or non-specific effects.
The presence of bovine serum albumin (BSA) as a carrier protein in recombinant rhesus macaque TNF-alpha preparations can significantly impact experimental outcomes:
Benefits of carrier protein:
Enhances protein stability by preventing adsorption to surfaces
Increases shelf-life by protecting against degradation
Allows the recombinant protein to be stored at more dilute concentrations
Maintains biological activity during freeze-thaw cycles
Potential experimental implications:
Immunological assays: BSA can interfere with antibody-based detection methods, especially when:
Using anti-bovine antibodies that might cross-react with BSA
Developing immunoassays where BSA might compete for binding
Performing immunoprecipitation experiments
Cell culture applications:
BSA may contain bioactive contaminants that affect cellular responses
For serum-free culture systems, BSA might introduce undefined factors
In low-serum conditions, carrier BSA might affect baseline cellular functions
Protein interaction studies:
BSA can non-specifically bind to other proteins
May interfere with protein-protein interaction studies
Could affect surface plasmon resonance or other binding assays
When to use carrier-free preparations:
The carrier-free version (without BSA) is recommended for applications in which the presence of BSA could interfere , including:
Mass spectrometry-based analyses
Crystallization studies
Certain receptor binding assays
Development of diagnostic tests
Experiments requiring precise protein quantification
Control strategies:
Include BSA-only controls at equivalent concentrations
Compare results between carrier-containing and carrier-free preparations
Include appropriate blocking steps in immunoassays to minimize BSA interference
Consider the final BSA concentration in the experimental system
By understanding these considerations, researchers can select the appropriate TNF-alpha preparation format and implement necessary controls to ensure valid experimental outcomes.
Validating the specificity of biological responses to recombinant rhesus macaque TNF-alpha is crucial for ensuring reliable and interpretable results:
Receptor Blocking Experiments:
Use neutralizing antibodies against TNF receptors (TNFR1 and TNFR2)
Pretreat cells with these antibodies before adding recombinant TNF-alpha
A significant reduction in TNF-alpha effects confirms receptor-specific action
Include isotype control antibodies as negative controls
TNF-alpha Neutralization:
Use anti-TNF-alpha neutralizing antibodies
Preincubate recombinant TNF-alpha with these antibodies before addition to cells
Abolishment of the biological effect confirms specificity
Include non-specific antibodies as controls
Dose-Response Relationships:
Perform detailed dose-response experiments
Plot the relationship between TNF-alpha concentration and biological response
A sigmoidal dose-response curve with EC50 values in the expected range (pg/mL to ng/mL) supports specificity
Unusual dose-response patterns may indicate non-specific effects
Pathway-Specific Inhibitors:
Use inhibitors of TNF signaling pathways (e.g., NF-κB inhibitors)
If these block the effects of recombinant TNF-alpha, this supports pathway-specific action
Include appropriate vehicle controls
Cross-Species Validation:
Compare the effects of rhesus macaque TNF-alpha with human TNF-alpha
Similar patterns of biological activity but potentially different potencies would be expected based on the 97% sequence identity
Include species-specific controls when possible
Biological Activity Profile:
Test multiple biological activities associated with TNF-alpha (cytotoxicity, cytokine induction, NF-κB activation)
A consistent pattern of activity across different assays supports specificity
Include positive and negative controls in each assay
By employing multiple validation approaches, researchers can confidently establish the specificity of biological responses to recombinant rhesus macaque TNF-alpha.
Recombinant rhesus macaque TNF-alpha serves as a valuable tool in infectious disease research, particularly for studies using non-human primates as models for human diseases:
HIV/SIV Research:
Study TNF-alpha's role in immune activation during chronic infection
Investigate interactions between viral proteins and TNF signaling pathways
Evaluate TNF-alpha as a biomarker for disease progression
Test anti-TNF strategies as adjunctive therapies
Tuberculosis Models:
Examine TNF-alpha's role in granuloma formation and maintenance
Study the impact of TNF blockade on TB reactivation
Investigate host-directed therapies targeting TNF signaling
Assess TNF-alpha as a biomarker for treatment response
Malaria Research:
Investigate the role of TNF-alpha in pathogenesis of severe malaria
Study the relationship between TNF-alpha polymorphisms and disease susceptibility
Test anti-TNF interventions to reduce immunopathology
Examine TNF-alpha's role in developing protective immunity
Viral Hemorrhagic Fevers:
Study TNF-alpha's contribution to vascular leak syndrome
Evaluate TNF-alpha as a biomarker for disease severity
Test anti-TNF strategies to reduce immunopathology
Investigate cytokine networks during infection
Experimental Approaches:
Ex vivo stimulation: Use recombinant TNF-alpha to stimulate immune cells from infected animals
In vivo administration: Administer TNF-alpha to study systemic effects
Neutralization studies: Use anti-TNF antibodies to block endogenous TNF-alpha
Co-culture systems: Study interactions between infected cells and TNF-alpha-producing cells
The 97% sequence identity between rhesus and human TNF-alpha makes rhesus macaque models particularly valuable for translational research on infectious diseases that are difficult to study directly in humans.
When using recombinant rhesus macaque TNF-alpha in inflammation and autoimmunity studies, several critical factors must be considered to ensure valid and translatable results:
Dosing and Pharmacokinetics:
Determine physiologically relevant doses based on measured levels in disease states
Consider the half-life of recombinant TNF-alpha in experimental systems
Develop appropriate dosing schedules for chronic versus acute inflammation models
Account for differences in local versus systemic administration
Receptor Specificity and Distribution:
Consider the differential expression of TNFR1 (ubiquitous) versus TNFR2 (hematopoietic-restricted)
Account for potential differences in receptor distribution between tissues
Remember that only TNFR1 contains a cytoplasmic death domain triggering apoptosis
Consider the impact of soluble TNF receptors that can neutralize TNF-alpha activity
Genetic Background Considerations:
Be aware of TNF-alpha promoter polymorphisms in different macaque populations
Consider how genetic background might influence TNF-alpha responsiveness
Ensure consistent genetic backgrounds within experimental groups
Account for potential differences between Chinese-origin versus Indian-origin rhesus macaques
Model-Specific Factors:
For colitis models: consider TNF-alpha's effects on intestinal barrier function
For arthritis models: account for TNF-alpha's effects on synoviocytes and osteoclasts
For psoriasis models: consider TNF-alpha's impact on keratinocyte proliferation
For multiple sclerosis models: account for TNF-alpha's dual roles in CNS inflammation
Experimental Design Considerations:
Include appropriate timing for TNF-alpha administration relative to disease induction
Consider combination treatment with other cytokines that synergize with TNF-alpha
Account for potential compensatory mechanisms with chronic TNF-alpha exposure
Include adequate controls for carrier proteins and endotoxin contamination
Translational Relevance:
Compare findings to human studies whenever possible
Consider the impact of anatomical and physiological differences between species
Evaluate how differences in immune system development might affect outcomes
Account for differences in environmental exposures between laboratory animals and humans
By carefully considering these factors, researchers can maximize the translational value of studies using recombinant rhesus macaque TNF-alpha in inflammation and autoimmunity research.